the integrity of a lamin-b1-dependent nucleoskeleton is a ... · organisation, rna synthesis,...

11
1014 Research Article Introduction Over the past 20 years, nuclear organisation in higher eukaryotes has been discussed at length (Jackson, 1991; Spector, 1993; Berezney et al., 1996; Cook, 1999; Misteli, 2004), and it is generally agreed that nuclear structure and function are inextricably linked. If this is true, it is also true that remarkably little is known about the molecular principles by which the links are made. A key focus of this debate has been the possible role of a unifying framework – the nucleoskeleton or nuclear matrix – that spreads throughout the nucleus. To date, however, although compelling evidence for such a structure continues to accumulate, a convincing molecular characterisation has remained elusive. In mammalian cells, the nuclear lamin proteins are major candidates to perform any structural roles; they form a structural nuclear framework that is composed of a peripheral nuclear lamina and a diffuse internal network (Goldman et al., 2002; Gruenbaum et al., 2003; Broers et al., 2006), with associated actin and myosin (Goldman et al., 2002; de Lanerolle et al., 2005). Two classes of lamin proteins, the A- and B-type lamins, are expressed from three genes – LMNA, LMNB1 and LMNB2. Somatic cells express at least one B-type lamin, and A-type lamins are expressed in differentiated cells, with LMNA expressing two major A-type lamins – A and C. Genetic alterations in human laminopathies (Worman and Courvalin, 2005; Broers et al., 2006) and transgenic mice with different patterns of lamin gene expression (Mounkes et al., 2003; Gruenbaum et al., 2003) demonstrate that a major function of the lamin proteins is to provide nuclear strength and integrity. In addition, lamin filaments are known to interact with many proteins, and defects in these interactions contribute to a variety of disease phenotypes (Worman and Courvalin, 2005; Broers et al., 2006). It is widely reported that lamin proteins contribute to the normal regulation of DNA and RNA synthesis, and it has been suggested that ‘lamins form a scaffold required for proper organisation of both replication and transcription’ (Goldman et al., 2002). Indeed, key published studies supporting this view have been performed using pseudo-nuclei assembled in vitro using Xenopus egg extracts (reviewed by Broers et al., 2006). To date, crucial roles for the nucleoskeleton in DNA and RNA synthesis of mammalian cells are based largely on circumstantial evidence. For example, one preliminary report suggests that disrupting the lamin network in BHK 21 cells will inhibit pre-mRNA synthesis (Spann et al., 2002), although a detailed characterisation of the transcription phenotype was not performed. Other observations suggest that assembled lamin proteins are not of fundamental importance in RNA synthesis. Studies of transgenic mice that are null for lamin A/C (Sullivan et al., 1999) or express a mutated form of lamin B1 (Vergnes et al., 2004) appear to be decisive. The lamin A/C knockout mice display complex phenotypes and although viable die prematurely. Mice expressing the mutant lamin B1 die at birth from respiratory failure. Yet the ability to generate embryonic fibroblasts from these lamin-modified mice implies that neither lamin A/C nor B1 are essential for cell growth and proliferation. This conclusion does conflict, however, with studies using RNA interference (RNAi) to deplete lamin expression, which show that B-type but not A-type lamins are required for cell proliferation (Harborth et al., 2001). Spatial organisation of nuclear compartments is an important regulator of chromatin function, yet the molecular principles that maintain nuclear architecture remain ill-defined. We have used RNA interference to deplete key structural nuclear proteins, the nuclear lamins. In HeLa cells, we show that reduced expression of lamin B1, but not lamin A/C, severely inhibits RNA synthesis – first by RNA polymerase II and later by RNA polymerase I. Declining levels of transcription correlate with different morphological changes in major nuclear compartments, nucleoli and nuclear speckles. Ultimately, nuclear changes linked to the loss of synthetic activity result in expansion of the inter-chromatin domain and corresponding changes in the structure and spatial organisation of chromosome territories, which relocate towards the nuclear periphery. These results show that a lamin B1-containing nucleoskeleton is required to maintain RNA synthesis and that ongoing synthesis is a fundamental determinant of global nuclear architecture in mammalian cells. Supplementary material available online at http://jcs.biologists.org/cgi/content/full/121/7/1014/DC1 Key words: Nuclear lamin proteins, Nucleoskeleton, Nuclear organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is a fundamental determinant of RNA synthesis in human cells Chi W. Tang 1, *, Apolinar Maya-Mendoza 1, *, Catherine Martin 1 , Kang Zeng 1 , Songbi Chen 1 , Dorota Feret 2 , Stuart A. Wilson 3 and Dean A. Jackson 1,‡ 1 Faculty of Life Sciences, University of Manchester, MIB, 131 Princess Street, Manchester, M1 7DN, UK 2 Cancer Research UK, Paterson Institute for Cancer Research, Christie Hospital, Wilmslow Road, Manchester, M20 9BX, UK 3 Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK *These authors contributed equally to this work Author for correspondence (e-mail: [email protected]) Accepted 9 January 2008 Journal of Cell Science 121, 1014-1024 Published by The Company of Biologists 2008 doi:10.1242/jcs.020982 Journal of Cell Science

Upload: others

Post on 03-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1014 Research Article

IntroductionOver the past 20 years, nuclear organisation in higher eukaryoteshas been discussed at length (Jackson, 1991; Spector, 1993;Berezney et al., 1996; Cook, 1999; Misteli, 2004), and it isgenerally agreed that nuclear structure and function are inextricablylinked. If this is true, it is also true that remarkably little is knownabout the molecular principles by which the links are made. A keyfocus of this debate has been the possible role of a unifyingframework – the nucleoskeleton or nuclear matrix – that spreadsthroughout the nucleus. To date, however, although compellingevidence for such a structure continues to accumulate, a convincingmolecular characterisation has remained elusive.

In mammalian cells, the nuclear lamin proteins are majorcandidates to perform any structural roles; they form a structuralnuclear framework that is composed of a peripheral nuclear laminaand a diffuse internal network (Goldman et al., 2002; Gruenbaumet al., 2003; Broers et al., 2006), with associated actin and myosin(Goldman et al., 2002; de Lanerolle et al., 2005). Two classes oflamin proteins, the A- and B-type lamins, are expressed from threegenes – LMNA, LMNB1 and LMNB2. Somatic cells express at leastone B-type lamin, and A-type lamins are expressed in differentiatedcells, with LMNA expressing two major A-type lamins – A and C.Genetic alterations in human laminopathies (Worman and Courvalin,2005; Broers et al., 2006) and transgenic mice with different patternsof lamin gene expression (Mounkes et al., 2003; Gruenbaum et al.,2003) demonstrate that a major function of the lamin proteins is toprovide nuclear strength and integrity. In addition, lamin filamentsare known to interact with many proteins, and defects in these

interactions contribute to a variety of disease phenotypes (Wormanand Courvalin, 2005; Broers et al., 2006).

It is widely reported that lamin proteins contribute to the normalregulation of DNA and RNA synthesis, and it has been suggestedthat ‘lamins form a scaffold required for proper organisation of bothreplication and transcription’ (Goldman et al., 2002). Indeed, keypublished studies supporting this view have been performed usingpseudo-nuclei assembled in vitro using Xenopus egg extracts(reviewed by Broers et al., 2006). To date, crucial roles for thenucleoskeleton in DNA and RNA synthesis of mammalian cells arebased largely on circumstantial evidence. For example, onepreliminary report suggests that disrupting the lamin network inBHK 21 cells will inhibit pre-mRNA synthesis (Spann et al., 2002),although a detailed characterisation of the transcription phenotypewas not performed.

Other observations suggest that assembled lamin proteins are notof fundamental importance in RNA synthesis. Studies of transgenicmice that are null for lamin A/C (Sullivan et al., 1999) or expressa mutated form of lamin B1 (Vergnes et al., 2004) appear to bedecisive. The lamin A/C knockout mice display complex phenotypesand although viable die prematurely. Mice expressing the mutantlamin B1 die at birth from respiratory failure. Yet the ability togenerate embryonic fibroblasts from these lamin-modified miceimplies that neither lamin A/C nor B1 are essential for cell growthand proliferation. This conclusion does conflict, however, withstudies using RNA interference (RNAi) to deplete lamin expression,which show that B-type but not A-type lamins are required for cellproliferation (Harborth et al., 2001).

Spatial organisation of nuclear compartments is an importantregulator of chromatin function, yet the molecular principlesthat maintain nuclear architecture remain ill-defined. We haveused RNA interference to deplete key structural nuclearproteins, the nuclear lamins. In HeLa cells, we show thatreduced expression of lamin B1, but not lamin A/C, severelyinhibits RNA synthesis – first by RNA polymerase II and laterby RNA polymerase I. Declining levels of transcription correlatewith different morphological changes in major nuclearcompartments, nucleoli and nuclear speckles. Ultimately,nuclear changes linked to the loss of synthetic activity result inexpansion of the inter-chromatin domain and corresponding

changes in the structure and spatial organisation of chromosometerritories, which relocate towards the nuclear periphery. Theseresults show that a lamin B1-containing nucleoskeleton isrequired to maintain RNA synthesis and that ongoing synthesisis a fundamental determinant of global nuclear architecture inmammalian cells.

Supplementary material available online athttp://jcs.biologists.org/cgi/content/full/121/7/1014/DC1

Key words: Nuclear lamin proteins, Nucleoskeleton, Nuclearorganisation, RNA synthesis, Chromosome territories

Summary

The integrity of a lamin-B1-dependent nucleoskeletonis a fundamental determinant of RNA synthesis inhuman cellsChi W. Tang1,*, Apolinar Maya-Mendoza1,*, Catherine Martin1, Kang Zeng1, Songbi Chen1, Dorota Feret2,Stuart A. Wilson3 and Dean A. Jackson1,‡

1Faculty of Life Sciences, University of Manchester, MIB, 131 Princess Street, Manchester, M1 7DN, UK2Cancer Research UK, Paterson Institute for Cancer Research, Christie Hospital, Wilmslow Road, Manchester, M20 9BX, UK3Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK*These authors contributed equally to this work‡Author for correspondence (e-mail: [email protected])

Accepted 9 January 2008Journal of Cell Science 121, 1014-1024 Published by The Company of Biologists 2008doi:10.1242/jcs.020982

Jour

nal o

f Cel

l Sci

ence

Page 2: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1015Linking nuclear structure and function

With particular emphasis on understanding the interplay betweennuclear structure and function, we have performed a detailedanalysis of the consequences of lamin protein depletion. UsingRNAi to reduce the expression of the nuclear lamin proteins, weshow that depletion of the major B-type lamin – lamin B1 – resultsin a specific inhibition of RNA synthesis. Loss of functional capacityis accompanied by profound changes in nuclear organisation, withdecreased RNA synthesis correlating with collapse of activechromatin and expansion of the inter-chromatin compartment. Asa consequence of this structural reorganisation, chromosometerritories (CTs) accumulate at the nuclear periphery. This suggeststhat the organisation of CTs is maintained by the association ofchromatin with active centres of RNA synthesis and that the activityof these centres is dependent on a nuclear structure that incorporateslamin B1.

ResultsDisrupting the nucleoskeleton in mammalian cellsRNAi was used to reduce expression of lamin proteins in humancells using both vector-based systems and direct delivery of shortinterfering RNAs (siRNA). Initially, RNAi was performed using avector-based protocol with co-transfection of vectors expressing asuitable reporter gene to identify individual RNAi-treated cells.Following RNAi, quantitative image analysis showed that selectedRNAi targets reduced expression of the appropriate lamin proteinto ~20% of the endogenous levels at 36-48 hours after transfection(Fig. 1). Further reduction was seen between 48 and 72 hours.

Depletion of lamin B1 inhibits RNA synthesisWe first evaluated how altered lamin geneexpression influenced RNA synthesis. Sites of pre-mRNA synthesis were visualised using an antibody(H5) that binds to the active form of RNApolymerase II and hence marks active transcriptionfactories (Fig. 2). Cells were co-transfected withRNAi vector and vector expressing YFP-tubulin.After different times, cells were fixed andtranscription sites visualised by indirectimmunofluorescence (Fig. 2A). Quantitative imageanalysis (Fig. 2C) showed that the amount of activeRNA polymerase II fell to ~20% of control levelsby 48 hours after transfection, with a progressiveloss of active RNA polymerase II seen between 12and 48 hours following transfection (supplementarymaterial Fig. S1). Cells with reduced expression of

lamin A/C (Fig. 2A) showed no reduction in active RNA polymeraseII. In parallel samples, RNAi was also performed with co-transfection of a vector that expresses an RNAi immune variant ofhuman lamin B1 gene (Materials and Methods), to counter thepossibility of protocol-dependent artefacts such as off-target effects.Cells expressing this modified LMNB1 showed no loss of activeRNA polymerase II (Fig. 2A).

To confirm reduced transcription, we also visualised nascent RNAdirectly after incorporating BrUTP (Fig. 2B). Following RNAitreatment, cells were permeabilised and nascent centres pulse-labelled with BrUTP in vitro. Analysis of cells with reduced laminB1 showed that the activity of RNA polymerase II was particularlysensitive to levels of lamin B1 expression, with pre-mRNA synthesisfalling to as little as 10% of control levels in cells, with <20% oflamin B1 found in controls (Fig. 2B,E). Cells with reduced laminB1 expression showed a clear loss in pre-mRNA synthesis, despitethe broad scatter in control cells. This cell-to-cell variability in RNAsynthesis and polymerised lamin B1 result from natural variabilitiesin HeLa cells, such as variable DNA content and changes duringthe cell cycle – as the number of active genes doubles as the genomeis replicated during S phase. The relationship between assembledlamin B1 and pre-mRNA synthesis suggests a direct link betweentranscription and the integrity of lamin-B1-containing nuclearstructures.

Cells with lamin B1 expression of 10-20% of controls alsoshowed declining levels of pre-rRNA synthesis (Fig. 2B), whicheventually resulted in the collapse of nascent pre-ribosomal

Fig. 1. Inhibiting expression of nuclear lamin proteins usingRNAi. RNAi was used to inhibit lamin protein expression inHeLa cells and analysis was performed on individual cells.(A,B) RNAi was performed by transient expression andtransfection monitored by co-transfection of a vectorexpressing YFP-tubulin (green). At selected times aftertransfection (48 hours in this example) cells were fixed andimmunolabelling of the residual lamina performed (red).Histograms (C) show averaged data (fluorescent intensitiesexpressed as arbitrary units; n>75) from three equivalentexperiments in which the average expression of lamin B1and A/C was reduced (KD) to 20.7 and 19.8% ofuntransfected cells (Control) in the same samples. Asnegative controls, cells were transfected with empty vector(not shown) or appropriate scrambled RNAi (C). Scale bars:10 μm.

Jour

nal o

f Cel

l Sci

ence

Page 3: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1016

transcripts into a small number of large nucleolar foci(supplementary material Fig. S2). These progressive changes areemphasised by the relative activities of the two classes of nascenttranscript (Fig. 2D). As expected, the ratio of pre-rRNA/pre-mRNAsynthesis in control cells was ~1.2; ~55% of nascent synthesis isnucleolar in HeLa cells. As lamin B1 expression fell, an initial fallof pre-mRNA synthesis and subsequent decay of pre-rRNAsynthesis resulted in a ~fourfold increase in the synthetic ratio. Aspredicted from the presence of active RNA polymerase II (Fig. 2A),depletion of laminA/C had no significant effect on the extent ofmRNA synthesis (Fig. 2F and supplementary material Fig. S3).

With vector-based RNAi, reporter gene expression provides anunambiguous assignment of treated cells, and untreated cells in thesample provide perfect experimental controls. However, becausesuch mixed cell populations are not suitable for biochemicalanalysis, we also performed RNAi using direct transfection ofsiRNA under conditions in which target gene expression is reduced

Journal of Cell Science 121 (7)

in >90% of cells (supplementary material Fig. S4). Importantly, thisconfirms that the average depletion of lamin proteins is very similarwhen quantitative immunofluorescence and immunoblotting areused (Figs 1, 2 and supplementary material Fig. S2); slightdifferences reflect the natural cell-to-cell variability of the vector-based approach. Using siRNA, a clear and progressive decrease inthe active form of RNA polymerase II, in transcription factories,was seen in cells with reduced lamin B1 expression, whereasdepletion of lamin A/C had no obvious effect.

Changes in global nuclear organisation in lamin-B1-depletedcellsWe next analysed how conditions of lamin B1 depletion, whichcompromise RNA synthesis, influenced markers of global nuclearorganisation, using well-characterised structural features of nucleoli(Raska et al., 2006) and nuclear speckles (Lamond and Spector,2003). The specific purpose of these experiments was to map the

Fig. 2. Reduced lamin B1 expression correlates with inhibition of RNA synthesis. HeLa cells were co-transfected with RNAi vectors and a vector expressing YFP-tubulin and the structure and activity of transcription sites monitored 48 hours later using either indirect immunofluorescence of the active form of RNApolymerase II (A) or BrUTP incorporation in permeabilised cells (B). (A) RNA polymerase (red) in transfected (expressing YFP-tubulin – green) cells aftertransfection with: lamin B1 RNAi vector (Kd LB1); RNAi vector co-transfected with lamin B1 rescue vector (Kb LB1 + Rescue); rescue vector alone (Rescuevector) and lamin A/C RNAi vector (Kd LA/C). (B) Nascent transcripts (BrU – red) in control cells and cells transfected with lamin A/C (Kd LA/C) and lamin B1(Kd LB1) RNAi vectors. Lamin B1 depletion was assessed by indirect immunofluorescence (Anti-LB1, red). As a control for possible labelling artefacts, the orderof antibody labelling was also reversed (Anti-LB1/BrU – merged image of lamin B1 labelled green and transcripts labelled red). Quantitative image analysis ofactive RNA polymerase II – H5 epitope (C) and nascent transcripts (D-F) in control and lamin-depleted cells showed that transcription was unaffected in cells withlamin A/C depleted (D,F) whereas depletion of lamin B1 (C-E) correlated with a clear decrease in transcription of both pre-mRNA and pre-rRNA (seesupplementary material Fig. S2). For pre-mRNA synthesis, statistical analysis using two-tailed Student’s t test showed the lamin-B1-depleted and control cellpopulations (E) to be unrelated. Scale bars: 10 μm (A); 5 μm (B).

Jour

nal o

f Cel

l Sci

ence

Page 4: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1017Linking nuclear structure and function

progressive changes in nuclear organisation that result fromdepletion of lamin protein expression. Analysis of the timing ofspecific changes would then give information about their temporalrelationship, and hopefully reveal how different defects werecausally related.

In these experiments, chromatin organisation was monitored byco-expression of DsRed-histone H2B during lamin depletion – notethat in these experiments the DsRed-histone is expressed andassembled into chromatin before cellular defects arise. Duringinterphase (Fig. 3A), the majority (~80%) of HeLa cells contained1-3 roughly spherical nucleoli, usually with diameters of 2-4 μm.Typically, each nucleolus had 10-20 active centres of ribosomalRNA synthesis, which are rich in the pre-rRNA processing proteinfibrillarin. Fibrillarin staining is used to monitor nucleolar activityas the complexity of the fibrillarin-stained active centres reflectsthe extent of rRNA synthesis – cells with active rRNA transcriptionhave multiple nucleoli with numerous small active sites, whereasfewer much larger sites are seen when transcription falls. About15% of proliferating cells had extended, ovoid-shaped nucleoli,which often touched the nuclear periphery. Very few cells containedlarge peri-nucleolar aggregates of fibrillarin or fibrillarin dispersedthroughout the cytoplasm (Fig. 3A). Depletion of lamin B1correlated with a >2.5fold fall in the most prominent interphaseorganisation and concomitant increase in the normally minororganisational patterns (Fig. 3A, histogram). These changescorrespond with structural changes seen in sites of pre-rRNAlabelling when RNA polymerase I activity is inhibited as a resultof >90% reduction in expression of lamin B1 (Fig. 2 andsupplementary material Fig. S2).

Nuclear speckles are nuclear domains of about 1 μm diameterthat are rich in proteins involved in pre-mRNA metabolism, suchas RNA splicing. The proteins within speckles are known to behighly dynamic, and this is reflected in the texture of the sites, whichis irregular and dispersed in transcriptionally active cells andspherical and condensed when transcription is suppressed (Lamondand Spector, 2003). The majority (~80%) of interphase HeLa cellscontained 10-20 irregular nuclear speckles, which were intenselystained by indirect imunolabelling using antibodies to the splicingprotein SC-35 (Fig. 3B). Almost all the other interphase cellscontained speckles with rounded and condensed appearance.Depletion of lamin B1 correlated with a >threefold fall in theprominent speckle pattern and increase in previously minor patterns,with the condensed speckle morphology increased from ~20 to~60% and a >fivefold increase in cells with diffuse nuclear andcytoplasmic labelling (Fig. 3B). These structural changes in themorphology of the nuclear speckle compartment are consistent firstwith a loss of the normal speckle structure, which mimics that seenwhen pre-mRNA synthesis is inhibited, and second with a generalloss of the nuclear targets that normally define the spatialorganisation of nuclear speckles in these cells.

During the course of these experiments we noticed that at latetime points (48-72 hours) progressively more cells developedatypical patterns of chromatin distribution (Fig. 3C). In nuclei ofcontrol cells, expression of DsRed-histone H2B defined theclassical amorphous chromatin distribution, with regions of denseand more dispersed chromatin surrounding chromatin-depletednuclear regions, which contain nucleoli and the interchromatinconstituents, such as nuclear speckles. This fenestrated appearancewas maintained following depletion of lamin A/C. However, insome cells loss of lamin B1 correlated with a profound alterationin nuclear organisation, with clear condensation of the dispersed

chromatin and expansion of the inter-chromatin compartment (Fig.3C).

Changes seen in cells with reduced lamin B1 expression donot arise through apoptosisStructural changes seen in lamin-B1-depleted cells at late time pointshave some features in common with nuclear changes that occurduring programmed cell death, although we also note that somechanges seen in nuclear organisation, particularly within thenucleolar compartment, are never seen during apoptosis. In viewof this, we next measured apoptosis in lamin-depleted cells usingAnnexin V binding (Fig. 4). Treating HeLa cells for only 4 hourswith staurosporin provided a positive control, with 100% of cellsshowing robust Annexin V binding (Fig. 4A). In negative controlsand cells treated to deplete lamin A/C ~2% of cells were Annexin-V-positive at 48 hours post-transfection and ~10% at 96 hours post-transfection (Fig. 4B). Notably, cells transfected with the lamin B1RNAi vector showed larger populations of Annexin-V-positive cells– with ~5 and 50% positive cells at 48 and 96 hours post-transfection, respectively (Fig. 4B). Although this shows thatdepletion of lamin B1 effects cell viability, consistent with earlierobservations (Harborth et al., 2001), the temporal changes in nuclearorganisation show that apoptosis is a consequence of events thatresult from depletion of lamin B1 and is inconsistent with thesuggestion that effects seen in lamin-B1-depleted cells arise becauseof some indirect mechanism that leads to apoptosis.

Phenotype of lamin depletion in other human cell typesThe status of p53 in HeLa cells is partially compromised byexpression of the HPV E6/7 proteins. To rule out any affects thismight have on apoptosis, we also performed lamin-depletionexperiments in primary lung fibroblast cells (MRC5) and colorectalcells (HTC116) with normal or null p53 status. In all cases, analysisof nascent RNA showed that these three cell types had the samedepletion phenotypes as HeLa cells when siRNAs were used toinhibit lamin protein expression (supplementary material Figs S5and S6). Additionally, the same results were seen when depletionexperiments were performed in cells treated with the apoptosisinhibitor Z-VAD-FMK (supplementary material Fig. S6).

Lamin B1 expression is required to maintain the organisationof chromosome territoriesChanges in chromatin organisation seen during lamin B1 depletion(Fig. 3C) prompted us to evaluate the organisation of CTs in thesecells. We first looked at a specific case using fluorescence in situhybridisation (FISH) to chromosome 19, which is normally locatedwithin the nuclear interior (Croft et al., 1999). Intriguingly, in lamin-B1-depleted cells this CT was seen to be localised at the nuclearperiphery. Notably, the nuclei of lamin-B1-depleted cells werefrequently disfigured during the FISH protocol so that the resultingsignals were more dispersed, and apparently weaker, than incontrols – consistent with depletion of lamin B1 compromisingglobal nuclear structure.

As the texture of DNA foci within CTs is lost during FISH, wenext imaged individual territories after labelling with BrdU. In thisexperiment, cell populations were grown in medium supplementedwith BrdU so that all DNA was labelled and then grown for ~5days to reveal individual CTs. Using high-resolution 3D imaging,BrdU-labelled territories displayed a complex structure of finelydispersed DNA foci – a typical example is shown in Fig. 5B(control). Following depletion of lamin B1, a general loss of the

Jour

nal o

f Cel

l Sci

ence

Page 5: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1018

surface texture of CTs was seen, with individual DNA foci appearingto become more condensed and tightly packed (Fig. 5B). At thesame time, this loss of structure correlated with an increasedtendency to associate with the nuclear periphery. Finally, weconfirmed these observations in living cells, using CTs labelled withAlexa Fluor 488 dUTP (Fig. 5C). Strikingly, in the most extremecases, the Alexa-labelled CTs appeared to be smeared against thenuclear rim. In more typical examples, the general shape of CTswas preserved but an expanded interchromatin domain correlatedwith CTs concentrated towards the nuclear periphery (Fig. 5D).These observations suggest that interactions that define the naturaldistribution of CTs are degraded when the expression of lamin B1is reduced.

Dynamic properties of the nuclear lamina during lamindepletionAs lamin depletion leaves a residual lamina with compromisedstructural properties (Fig. 5), we next evaluated the stability of thenucleoskeleton in cells with depleted lamin expression. Inproliferating mammalian cells, soluble and lamina-associatedsubunits have t1/2 for exchange of >2 hours (Moir et al., 2000).Under growth conditions used here, the t1/2 for recovery of GFP-lamin A in HeLa was ~60 minutes (Fig. 6). When lamin A and B1were depleted, the t1/2 for recovery of GFP-lamin B1 or A of theresidual lamin fell to ~20 and 10 minutes, respectively. These dataconfirm that although the lamina is a stable structure, the naturalstability is dependent on a balance of lamin proteins that contributeto the structural network of the intact lamina.

These changes in the behaviour of the residual lamina raiseobvious questions about its structure and any complementationeffects that are seen in lamin-depleted cells. In agreement with theanalysis of Lammerding et al. (Lammerding et al., 2006), we foundthat cells with depleted lamin protein expression showed verylimited upregulation of other lamin proteins. For example, usingvector-based depletion of lamin A/C the distribution of lamin B1was clearly normal using indirect immunofluorescence(supplementary material Fig. S2). Quantitative analysis, usingdepletion with siRNAs, showed that in all cases expression of theunaffected lamin proteins was between 100 and 120% of the normallevels (supplementary material Figs S4 and S5).

Analysing the chromatin loops of lamin-depleted cellsLamin-dependent nuclear structures are known to play a key rolein nuclear mechanics and shape determination (Broers et al., 2006;Lammerding et al., 2006). In addition, the nucleoskeleton has alsobeen implicated in the organisation of chromatin loops (Jackson,1991). With this in mind, we next evaluated the structure of DNAloops in nucleoids prepared from cells with reduced lamin geneexpression (Fig. 7). Lamin gene expression was reduced usingvector-based RNAi and co-expression of reporter genes that markthe nuclear rim (Fig. 7) and nucleoids treated with ethidiumbromide to monitor the structure of expanded nuclear halos (Fig.7). Notably, the DNA halos from lamin-A/C-depleted nuclei wereindistinguishable from untreated cells, whereas cells with reducedexpression of lamin B1 showed general loss of structure of theresidual nucleoid cage and corresponding deformities in the structureof the DNA halo (Fig. 7A). Specifically, in lamin-B1-depleted cells,DNA halos were more expanded than in controls but also muchmore irregular; in control as well as lamin-A/C-depleted cells haloswere generally spherical, whereas halos from lamin-B1-depletedcells were asymmetrical, with huge DNA bundles spreading many

Journal of Cell Science 121 (7)

microns away from the nucleoid cage. This loss of general looporganisation was particularly evident during time-lapse analysis,where the majority of lamin-B1-depleted cells showed profoundloss of nucleoid structure, often with huge plumes of DNA escapingfrom nucleoids under ultraviolet illumination (Fig. 7B). Theseobservations confirm that the integrity of the lamin-B1-containingnucleoskeleton is a major determinant of the organisation ofchromatin domains in HeLa cells.

DiscussionIn human cells, nuclear lamin proteins provide an intermediatefilament network that maintains nuclear structure (Broers et al.,2006). This network may also support other organisational roles.For example, it is possible that by organising nuclear actin andmyosin the lamin network will orchestrate spatial dynamicswithin the nucleus (Chuang et al., 2006). Nuclear lamin proteinshave also been reported to influence different aspects of nuclearfunction (reviewed by Broers et al., 2006; Gruenbaum et al., 2003).However, if lamin proteins do regulate fundamental functions, itis surprising that viable cells can be isolated from mice withdeficient LMNA (Sullivan et al., 1999) or LMNB1 (Vergnes et al.,2004) expression.

In the mouse models, deletion of lamin A/C expression – as anull phenotype – has similar properties to some classes of humanlaminopathies, with developmental defects particularly in muscle,heart and peripheral nerves, and the lamin-B1-defective mice dieat birth with severe developmental phenotypes (Vergnes et al., 2004).Even so, the ability to establish cell lines (MEFs) from embryos ofLMNB1 mutant mice implies that loss of normal lamin B1 functionis not inevitably cell lethal. However, these observations on murinecells do contradict RNAi experiments with human cells, whichsuggest that B-type but not A-type lamins are required for cellproliferation, with prolonged reduction in expression of B-typelamins resulting in apoptosis (Harborth et al., 2001).

To explain this conflict, it might be argued that specific functionsplayed by nuclear lamins are cell-context-specific. Even so, it isnotable that the LMNB1 mouse was generated using a recombinationstrategy that left the promoter intact to express a fusion protein inwhich the 3� half of the lamin B1 was replaced by the βgeo reporter(Vergnes et al., 2004). Despite the fact that the nuclear localisationsignal was deleted in this construct, the altered lamin B1 in MEFslocalises to the nucleus and concentrates at the nuclear periphery.This construction and the presence of a mutant form of lamin B1in the residual lamin complicates any conclusions that might bemade about the normal function of lamin B1. Clearly, LMNB1 MEFshave lamina with altered structural properties and compromiseddifferentiation potential (Vergnes et al., 2004), which might alsoexplain the observation that MEFs with altered lamin B1 expressiondisplay abnormalities in CT distribution (Malhas et al., 2007).However, these observations do not exclude the possibility that somelamin B1 functionality might be at least partially retained.

Adding to this controversy, a preliminary study from Goldmanand colleagues (Spann et al., 2002) provides compelling data tolink the organisation of the lamin network to RNA synthesis insomatic mammalian cells. Here, a dominant-negative knockoutstrategy – using N-terminally-deleted lamin A – was used to disruptthe lamin filaments. The loss of lamin A filaments was reported tocorrelate, qualitatively, with a decay in transcription by RNApolymerase II. However, the interpretation of this observation iscomplicated because the disruption also removes B-type laminsfrom the nuclear periphery to nuclear aggregates that sequester

Jour

nal o

f Cel

l Sci

ence

Page 6: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1019Linking nuclear structure and function

essential transcriptional factors, giving potentially profoundbystander effects.

We wanted to confirm possible roles for nuclear lamin proteinsin linking nuclear structure and function. To do this we used RNAistrategies that deplete individual lamin proteins without significantlyaltering the concentration of the remaining lamin proteins in theresidual lamina. We describe a detailed analysis of structure-functionrelationships in four human cell lines. In HeLa cells, depletion of

lamin A/C expression had no obvious phenotype, whereas depletionof the major B-type lamin – lamin B1 – correlated with a pronouncedloss of transcriptional activity. The same transcription defects wereseen in primary lung fibroblasts and colorectal cells with normaland null p53 status.

During lamin B1 depletion, levels of pre-mRNA synthesis fellconcomitantly with loss of lamin B1 (Fig. 2); active RNApolymerase was lost (Fig. 2), although overall expression of RNA

Fig. 3. Lamin B1 depletion correlates with altered morphology in nucleoli and nuclear speckles. Cells co-transfected with RNAi vectors and a vector expressinghistone H2B were analysed after 48 hours to monitor the morphology of major nuclear compartments. (A) Sites of pre-rRNA synthesis in nucleoli were visualisedby indirect immunolabelling of fibrillarin (green). Changes were monitored in cells expressing DsRed-histone H2B (red) and different patterns recorded; shown incartoon form and occurrence as a frequency histogram [LB1 kd, 187 cells (four experiments); Control, 100 cells (two experiments)]. Control cells were transfectedwith empty vector, although untransfected cells in RNAi samples also serve as controls. (B) Nuclear speckles were visualised by indirect immunolabelling of thesplicing factor SC-35 (green). Structural changes were monitored in cells expressing DsRed-histone H2B (red) and the appearance and frequency of major changesdescribed (LB1 kd, 196 cells (five experiments); Control, 98 cells (two experiments). (C) The structure of the chromatin compartment in HeLa cells was monitoredby expression of DsRed-histone H2B. Typical examples of untreated cells (control) and cells with lamin A/C or B1 depleted (Kd LA/C; Kd LB1) are shown. Scalebars: 5 μm (A,C); 10 μm (B).

Jour

nal o

f Cel

l Sci

ence

Page 7: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1020

polymerase II changed little (supplementary material Fig. S5). Theconsequence of this was seen in global changes in nuclearorganisation (Fig. 3). During the early stage of lamin B1 depletion,pre-rRNA synthesis in nucleoli was unaffected. However, at laterstages, pre-rRNA synthesis was inhibited and characteristic changesin nucleolar morphology seen (Figs 2, 3 and supplementary materialFig. S2). This is consistent with a lamin-B1-dependent nuclearstructure being required for assembly of active sites of RNAsynthesis. By contrast, reducing the level of lamin A/C had noobvious phenotype under the routine knockdown conditions used.This dominant behaviour of lamin B1 is surprising, as embryonicmouse fibroblasts with no lamin B1 are viable in culture (Vergneset al., 2004). A possible explanation for this is that the structuraland functional properties of the nucleoskeleton are defined by itsoverall composition, so that differential expression of the B-typelamins in different cells and tissues will allow some functionalcomplementation [see Lammerding et al. (Lammerding et al., 2006)for discussion of effects of complementation in cells from knockoutmice]. Even so, in experiments performed here, altered expressionof the remaining lamins was no more than 20% of the normal levelsand therefore unlikely to significantly compensate for any defectsseen. Additionally, in all four cells lines used, depletion of laminB1 but not lamin B2 resulted in transcriptional inhibition.

Depletion of lamin B1 is ultimately lethalConditions that activate programmed cell death in human cells areextremely complex. In the nucleus, checkpoint pathways monitorDNA integrity and activate apoptosis if DNA damage accumulates(Roos and Kaina, 2006). Activation of apoptosis correlates with anumber of diagnostic nuclear changes (Robertson et al., 2000), withmany nuclear proteins, including both A- and B-type lamins,targeted for caspase-dependent degradation. Well-characterisednuclear changes then follow.

Depleting lamin B1 in HeLa cells leads to a progressive loss ofRNA synthesis and eventually results in cell death (Fig. 4).

Journal of Cell Science 121 (7)

However, this is a late phenotype, and we stress that all functionaland structural observations (Figs 2-5) detailed here were made usingcells that were morphologically normal using bright-fieldmicroscopy and chromatin organisation. Changes in RNA synthesiswere first evident within 12 hours of inducing lamin B1 depletion,and a robust and predictable programme of changes was seenbetween 18 and 48 hours (Figs 2-3), at least 2 days before asignificant proportion of depleted cells were positive for AnnexinV binding (Fig. 4). The timing of onset of apoptosis is consistentwith cell death arising as a result of defects in gene expression, sothat cells become starved of components that are required to sustainessential cellular processes as levels of key transcripts fall.

This conclusion is supported by analysis of the effect of laminB1 depletion on DNA synthesis. Although our detailed analysis ofDNA synthesis (C.W.T., A.M.-M. and D.A.J., unpublished) fallsoutside the scope of this study, two key observations are worthy ofcomment. First, as for transcription, depletion of lamin B1 – butnot lamin A/C or B2 – gives rise to a replication phenotype (48-60 hours post-RNAi), which results in a ~50% decline of cells withinS phase but increase in the proportion of mid/late S-phase cells.Second, time-lapse analysis shows that cells with reduced laminB1 expression have frequent defects in the replication programme,including extended S-phase duration and abortion of synthesisbefore S phase is complete. It appears that S-phase-dependentdefects in cells with compromised lamin B1 expression will besufficient to activate replication checkpoints and induce apoptosis.

Lamin B1 links nuclear structure and functionMammalian nuclei are structured so that nuclear functions areperformed within dedicated nuclear sites (Cook, 1999). Chromatinthat interacts with these sites is locally structured, to give spatiallydefined CTs (Cremer and Cremer, 2001; Misteli, 2004), whichoccupy preferred nuclear positions that reflect transcriptionalactivity and differentiation status. However, as global molecularmechanisms that regulate the location of CTs have not been

Fig. 4. Monitoring apoptosis in lamin-depleted HeLa cells. HeLa cells with depleted lamin expression were analysed (48 or 96 hours after transfection) to monitorbinding of Annexin V. Cells were stained with Alexa-568 (red)-conjugated Annexin V and counterstained with Hoechst (blue) (A) and the percentage of Annexin-V-positive transfected cells (n=200) were determined (B). Cells on coverslips were treated as follows: (1) positive controls [Control (+)] treated with staurosporine (5μm in medium; 4 hours); (2) negative controls [Control (–)] transfected with pYFP-tubulin using polyfect; (3) lamin-A/C-depleted cells (KD LA/C) transfected withpYFP-tubulin and pSECpuroLaminA/C; and (4) lamin-B1-depleted cells (KB LB1) transfected with pYFP-tubulin and pSEChygroLaminB1. Scale bars: 10 μm.

Jour

nal o

f Cel

l Sci

ence

Page 8: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1021Linking nuclear structure and function

defined, the extent to which the position of a CT might influenceany aspect of function remains a matter for debate. Even so, recentstudies have suggested that the organisation of CTs could allowgenes that interact with similar regulatory transcription factors tobe expressed within common transcription factories (Chakalova etal., 2005; Spilianakis and Flavell, 2006), perhaps as independentlyregulated gene networks.

Our analysis shows that nuclear function, and specifically RNAtranscription, is a key determinant of chromosome position.Depletion of lamin B1 and corresponding loss of RNA synthesiscorrelates with redistribution of CTs towards the nuclear periphery(Fig. 5), mimicking changes seen in cells treated with inhibitors ofRNA synthesis (Croft et al., 1999). In addition, changes described

herein are reminiscent of structural changes described by Albiez etal. (Albiez et al., 2006) in cells grown under conditions of differingosmolarity. In this study, transferring cells to hypertonic mediainduced a dramatic but reversible collapse of chromatin andcorresponding expansion of the inter-chromatin domain. Hence, thecorrelation between chromatin organisation and function suggeststhat chromosome position will normally reflect the balance ofinteractions at the nuclear lamina and internal nucleoskeleton; abalance that is altered in lamin-B1-depleted cells.

Chromatin loops and the nucleoskeleton in human cellsOur observation that loss of lamin-B1-dependent structures in HeLaleads to reduced RNA synthesis implies that lamin-based structures

Fig. 5. Reduced lamin B1 expression correlates with altered location of chromosome territories. CTs were analysed in lamin-B1-depleted cells using: (A) FISH tochromosome 19 (wide-field imaging); (B) BrdU, by indirect immunofluorescence (high-resolution 3D imaging); (C) Alexa Fluor 488 dUTP, in living cells(confocal sections). (B,C) Unsynchronised cells were labelled and grown for 5-7 days so that most labelled cells retained 1-3 discrete, labelled territories. RNAivectors were transfected 48 hours before analysis. CTs were assigned to nuclear zones of equal area (Materials and Methods) using at least 100 territories/sample(A-C). Changes were analysed using two-tailed Student’s t test as follows: (A) control vs lamin A/C depletion (t=2.396, P<0.0172*); control vs lamin B1 depletion(t=9.203. P<1.567e-17***); (B) control vs lamin A/C depletion (t=0.478, P<0.634); control vs lamin B1 depletion (t=5.780, P<2.973e-8***); (C) control vs laminB1 depletion (t=6.442, P<1.996e-09***). Marginalisation of CTs (D, left panel, shows a single confocal section of CTs labelled with BrdU in green) correlatedwith collapse of open chromatin (D, centre panel, DsRed-histone H2B in red) and a corresponding expansion of the inter-chromatin compartment. Scale bars:10 μm (A); 5 μm (C,D).

Jour

nal o

f Cel

l Sci

ence

Page 9: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1022 Journal of Cell Science 121 (7)

function at transcription sites throughout the nuclear interior.However, although there is some evidence for an internalnucleoskeleton containing B-type lamins (Hozak et al., 1995;Barboro et al., 2002), the significance of this is difficult to assessas so little is known about the structure of the peripheral and internallamin networks (Gruenbaum et al., 2003; Broers et al., 2006).

Following lamin depletion, the dynamic behaviour of the residualproteins might hint at the structure of the network. Proteins of thenuclear lamina are known to exchange slowly. In proliferating cells,ectopically expressed fluorescent A- or B-type lamins exchangedwith a t1/2 in the range 1.5-3 hours (Moir et al., 2000; Gilchrist etal., 2004), although mutants with substantially increased mobilityhave been described (Gilchrist et al., 2004). In our hands, GFP-lamin A/C expressed ectopically in HeLa cells incorporated intothe endogenous lamina and exchanged with t1/2 of ~60 minutes(Fig. 6). A clear increase in the rate of exchange was seen wheneither A- or B-type lamins were depleted from the endogenouslamina, implying that both A- and B-type lamins contribute to thestability of the endogenous network.

Alteration in the structure of the lamin network was evident innucleoids prepared from lamin-B1-depleted cells, where loss ofstable DNA loops suggests that lamin-B1-dependent interactionsare a key determinant of chromatin loop organisation. Once again,these experiments emphasise a clear difference in the roles playedby A- and B-type lamins. While both classes of lamin protein appearto contribute to the structure of an integrated lamin protein network,the major roles of the two classes are distinct. Most notably, laminB1 is essential to maintain natural levels of gene expression, whereasloss of lamin A/C has no obvious functional implications. The majorrole of A-type lamins, by contrast, appears to be linked to thestructural maintenance of the nuclear compartment and regulationof nuclear mechanics (Lammerding et al., 2006).

In conclusion, we show here that lamin B1 is an essentialcomponent of a nuclear structure that is required to maintain thenatural activity of RNA synthesis in human cells. When lamin B1

is depleted, the decay of RNA synthesis also correlates withstructural alterations in major nuclear compartments, including CTs.We propose that the molecular network that connects a lamin-B1-dependent nucleoskeleton, nucleoskeleton-associated transcriptionfactories and chromatin templates, which interact with the activesites, provides a general molecular mechanism to explain hownuclear structure and function are linked in higher eukaryotes.

Materials and MethodsCell cultureHeLa cells (S3) and p53 normal and null human colorectal cells (HCT116) weregrown in DMEM supplemented with antibiotics and 10% FBS. Primary human lungfibroblast cells (MRC5) were grown in MEM supplemented with antibiotics and 10%FBS.

RNAi protocolsFor each target gene, three expressed target sequences were selected (Harborth et al.,2001) (www.ambion.com). RNAi was performed using double-stranded RNA(Ambion) or short hairpin RNAs expressed from pSuper. RNAi by duplex siRNAswas validated by western blotting and immunolabelling and the most effective targetscloned in to the RNAi pSilencerTM expression vectors, pSECpuro and pSEChygro(Ambion).

The following target sequences were chosen for detailed study: Lamin A/C,CTGGACTTCCAGAAGAACA; Lamin B1, AGAGTCTAGAGCATGTTTG; LaminB2, GCTGAGCTCTGACCAGAAC; B1-Scrambled, GTCGATAACGTGCTACTAT.

RNAi was performed on HeLa cells at 75% confluency in six-well plates. For eachwell, 1.5 μg plasmid DNA in 100 μl Optim medium without serum was mixed with12 μl Polyfect (Qiagen) and incubated for 5 minutes (20°C), after which time 0.6 mlfresh medium was added. Transfection mix was added to wells with 2 ml fresh medium(37°C). After 24 hours, cells were seeded on to 13 mm coverslips coated with poly-L-lysine. RNAi vectors were co-expressed with appropriate reporter genes, foridentification of the transfected cells. The reporters used were: YFP-tubulin (Clontech);DsRed- and GFP-lamin A/C and B1, DsRed-histone H2B. Plasmids with LMNA andLMNB1 were kind gifts from Chris Hutchison (Durham, UK) and Jan Ellenberg(EMBL, Germany), respectively. Cells were monitored for up to 96 hours followingtransfection and changes analysed by quantitative microscopy. Typically, 100-200cells were analysed for each sample.

Rescue of lamin B1 knockdown was performed using point mutations generatedby site-specific mutagenesis by overlap extension. The third nucleotides of each codonin the target sequence of lamin B1 cDNA were altered, to preserve the natural proteinsequence. Changes were made using a three-step PCR protocol with Roche ExpandHigh Fidelity PCR Kit and a 3� FLAG tag incorporated. Modified cDNA was cloned

Fig. 6. Stability of the nuclear lamina following modification by RNAi. HeLa cells were co-transfected with RNAi vectors and vector expressing the relevant GFP-laminA/C or B1 to monitor dynamics. After 48 hours, cells with low intensity reporter expression were selected and fluorescence recovery after photobleachingmonitored. Panels (A) show a typical FRAP profile (from left to right): pre-bleach image; image immediately after bleaching – in each cell analysed two 2 μmzones were bleached, one is shown boxed; and two frames (15 and 60 minutes) from a time-lapse series with images taken at 15 minute intervals over 2 hours.Analysis of bleach zones (B) was used to estimate t1/2 for fluorescent recovery for cells transfected with empty RNAi vector (Control) and cells with lamin B1(LB1 KD) and lamin A/C (LA/C KD) depleted. Data points shown in B are average intensities of ten bleach zones (five cells) for the long (300 minute) timecourseand 20 bleach zones (ten cells) for the short (50 minute) timecourse. Scale bars: 5 μm.

Jour

nal o

f Cel

l Sci

ence

Page 10: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

1023Linking nuclear structure and function

into PCEP4 vector (Invitrogen) to give PCEP4-lamin B1/RNAi–, with the modifiedlamin B1 cDNA expressed from the CMV promoter. Following transfection,expression of the RNAi immune lamin B1 protein was monitored by immunoblotting,using both anti-lamin B1 (Zymed) and anti-FLAG (Sigma). Lamin A/C siRNA (sc-

35776) and Lamin B1 siRNA (sc-29386) were purchased from Santa Cruz. LaminB2 siRNA and Scrambled siRNA were from Sigma-Genosys.

Labelling transcription factoriesTranscription factories were labelled by indirect immunofluorescence of the activeform of RNA polymerase II. Nascent transcripts were visualised followingincorporation of BrUTP in vitro (Jackson et al., 1993). Cells on coverslips werepermeabilised using Triton X-100 (0.1% for 1 minute at 20°C) and transcriptionperformed in buffer supplemented with 500 μM BrUTP, GTP, CTP and 2 mM ATP(4 minutes at 37°C).

For live imaging of DNA foci, cells were labelled with either Cy3-dUTP or AlexaFluor 488 dUTP for visualisation of DNA foci and CTs in living cells. Cells wereseeded on to 13 mm coverslips coated with poly-L-lysine and transfection performedat 50% confluency. For each coverslip, 3 μl Fugene6 was mixed with 1 μl dUTPanalogue (10 minutes; 0°C) and 17 μl PBS (further 5 minutes; 0°C) before applyingto cells (10 minutes; 0°C). Coverslips were rinsed and returned to fresh medium andseeded as required to visualise individual labelled CTs 4-7 days later.

Live cell imagingLive cell imaging was performed using a Zeiss LSM 510 META microscope.Fluorescence recovery after photobleaching (FRAP) was performed as follows. Cellsexpressing fluorescent lamin proteins were selected using laser scanning at low laserpower; RNAi-treated and control cells with similar, low level, expression were selectedfor comparison. A pre-bleach image was recorded (1-2% 488 laser line; two scansfor 1 μm confocal section) and a selected area then bleached using high laser power(full power for 488 and 543 lines). FRAP was recorded using time-lapse images ofthe original section and quantitative image analysis of the bleach zone performedusing the Zeiss software.

For live imaging of CTs, labelled cells were grown on 35 mm Petri dishes with13 mm glass inserts (MatTek Corporation) in medium without Phenol Red. Using a1.4NA 100� Plan-Apo DIC objective, cells were selected under bright-fieldillumination and then 3D image stacks taken under live scanning mode using minimumlaser power.

ImmunofluorescenceImmunofluorescence was performed under standard conditions (Pombo et al., 1999).Briefly, samples were rinsed in PBS, fixed with 4% paraformaldehyde (ElectronMicroscopy Sciences; 15 minutes on ice), blocked and immunolabelled using thereagents below. Slides were then washed, DNA stained with 5 μg/ml Hoechst 33258(Sigma) for 10 minutes, and mounted with Vectashield (Vector Laboratories). Thelamin antibodies used throughout this study were from Santa Cruz (Lamin A/C sc-7292, Lamin B2 sc-56147) and Abcam (lamin B1 ab6048).

Active RNA polymerase II in transcription factories – mouse anti-human RNApolymerase II antibody (Covance; H5 antibody – MMS-129R; 1/1000; 15 hours;4°C) and Cy3-donkey anti-mouse IgG (1/1000; 1 hour; 20°C).

BrdU detection in nascent RNA – sheep anti-BrdU antibody (Biodesign; M20105S;1/1000; 1 hour; 20°C) followed by Cy3- or Alexa-Fluor-488-conjugated donkey anti-sheep IgG (1/1000; 30 minutes; 20°C).

BrdU detection in DNA – after fixation, slides were rinsed twice in ddH2O andthen incubated with HCl (2.5 M; 1 hour; 20°C) to denature DNA. Slides were thenwashed three times in PBS, blocked and cells immunolabelled using sheep anti-BrdUantibody (Biodesign; M20105S; 1/1000; 1 hour; 20°C) and Cy3-donkey anti-sheepIgG (1/1000; 1 hour; 20°C).

Nucleoli – mouse monoclonal antibody to fibrillarin (Cytoskeleton – AFB01; 1/200;15 hours; 4°C) and FITC-conjugated donkey anti-mouse IgG (1:1000; 1 hour; 20°C).

Nuclear speckles – mouse monoclonal antibody to SC-35 (Sigma – S4045; 1/500;15 hours; 4°C) and FITC-conjugated donkey anti-mouse IgG (1:1000 dilution; 1 hour;20°C).

ApoptosisCells undergoing apoptosis were monitored by Annexin V binding. At appropriatetimes after transfection (typically 48 and 96 hours) cells were rinsed in PBS, fixedand stained with Annexin V conjugated directly with Alexa 568 (Roche – diluted1:50 in incubation buffer) as detailed by the manufacturer. Samples were washedextensively in PBS and nuclei counterstained with Hoechst 33258.

Caspase inhibition: 50 μM of Z-VAD-FMK (MBL International Corporation) wasadded into the medium the day before transfection.

FISHControl and RNAi-treated HeLa cells were grown on poly-L-lysine-coated slides andfixed with 4% paraformaldehyde (10 minutes at 20°C). The distribution ofchromosome 19 was determined using WCP 19 labelled with SpectrumGreenTM

(Vysis). FISH was performed as recommended by the manufacturer, with hybridisationfor 18 hours at 37°C.

Nuclear halo preparationsTo prepare nucleoids, HeLa cells were suspended in ice-cold PBS without Ca2+ andMg2+and aliquots of 50 μl containing 3.5�105 cells were gently mixed with 150 μl

Fig. 7. Changes in DNA loops in lamin-depleted cells. The structure of DNAhalos from lamin-depleted cells was analysed (A-C). Cells were depleted oflamin A/C or B1 using RNAi vectors and co-transfection with a vectorexpressing an appropriate fluorescent-lamin A/C or B1, as reporter that marksthe nuclear periphery. After 36 hours, nucleoids were prepared and samplescontaining ethidium bromide (A) used to define halo structure and thecorresponding DNA loop length (C). Time-lapse epi-fluorescence microscopywas also used to monitor the expansion of DNA halos, using 15 second timeframes over 2 minutes; two frames from typical time-lapse series at 30 and 90seconds are shown (B). DNA halos prepared from lamin A/C depleted cellsexpanded to give uniform halos of DNA loops of 19.1±3.9 μm (n=58), whichis equivalent to an average DNA loop, measured from the nuclear rim, of 112kbp; this was indistinguishable from the DNA loops of untransfected controlsin the same preparation (A). The average halo from lamn-B1-depleted cellswas 24.0±5.5 μm (n=64), equivalent to 141 kbp. White arrows highlightabnormal structures seen in lamin-B1-depleted cells. The size of scale bars isgiven on individual images. Scale bars: 20 μm.

Jour

nal o

f Cel

l Sci

ence

Page 11: The integrity of a lamin-B1-dependent nucleoskeleton is a ... · organisation, RNA synthesis, Chromosome territories Summary The integrity of a lamin-B1-dependent nucleoskeleton is

lysis solution containing 2.6 M NaCl, 1.3 mM EDTA, 2.6 mM Tris, 0.6% Triton X-100, pH 8.0. After 20 min at 4°C, the mixture was overlaid on sucrose step gradientscontaining 0.2 ml 30% sucrose under 0.6 ml 15% sucrose. Both sucrose layerscontained 2.0 M NaCl, 1.0 mM EDTA, 10 mM Tris, pH 8.0. The gradients werespun at 4°C in a microfuge for 4 minutes at 9000 g. Nucleoids (in ~250 μl buffer)were recovered, 50 μl stained with ethidium bromide to final concentration of 5 μg/mland examined by fluorescence microscopy. DNA halos expand continuously whenethidium-bromide-stained nucleoids are exposed to ultraviolet light. To monitor thestructure of the halos, it is therefore essential to use identical exposure times; in Fig.7, imaging was performed at 20 seconds.

Western blotWhole cell extracts (10 ng whole cell protein/lane) were used in all western blots.Actin anti-body was from Sigma-Aldrich (A1978). Goat anti-mouse HRP (172-1011)and goat anti-rabbit HRP (172-1019) were from Bio-Rad. Anti-RNA polymerase II,clone CTD4H8 was from Upstate. ECL Western Blot Analysis System (RPN2109)was from GE Healthcare.

Image processing and analysis3D images were generated from confocal Z series using Imaris software (Bitaplane,Switzerland). Centre of mass for chromosome territories (Fig. 5) was determined andassigned to specific nuclear sectors as described by Croft et al. (Croft et al., 1999).Statistical analysis was performed by linear regression and Student’s t test usingMicrosoft Excel.

We thank Chris Hutchison and Jan Ellenberg for providing reagentsthat were used in this study. This work was undertaken with financialsupport from BBSRC (C.W.T., A.M.-M., C.M., S.C., S.W., D.A.J.) andEuropean Commission (K.Z., D.F., D.A.J.).

ReferencesAlbiez, H., Cremer, M., Tiberi, C., Vecchio, L., Schermelleh, L., Dittrich, S., Kupper,

K., Joffe, B., Thormeyer, T., Cremer, T. et al. (2006). Chromatin domains and theinterchromatin compartment form structurally defined and functionally interacting nuclearnetworks. Chromosome Res. 14, 707-733.

Barboro, P., D’Arrigo, C., Diaspro, A., Mormino, M., Alberti, I., Parodi, S., Petrone,E. and Balbi, C. (2002). Unravelling the organisation of the internal nuclear matrix:RNA-dependent anchoring of NuMA to a lamin scaffold. Exp. Cell Res. 279, 202-218.

Berezney, R., Mortillaro, M. J., Ma, H., Wei, X. and Samarabandu, J. (1996). Thenuclear matrix: a structural milieu for genomic function. Int. Rev. Cytol. 162A, 1-65.

Broers, J. L. V., Ramaekers, F. C. S., Bonne, G., Ben Yaou, R. and Hutchison, C. J.(2006). Nuclear lamins: laminopathies and their role in premature ageing. Physiol. Rev.86, 967-1008.

Chakalova, L., Debrand, E., Mitchell, J. A., Osborne, C. S. and Fraser, P. (2005).Replication and transcription: shaping the landscape of the genome. Nat. Rev. Genet. 6,669-677.

Chuang, C. H., Carpenter, A. E., Fuchsova, B., Johnson, T., de Lanerolle, P. andBelmont, A. S. (2006). Long-range directional movement of an interphase chromosomesite. Curr. Biol. 16, 825-831.

Cook, P. R. (1999). The organization of replication and transcription. Science 284, 1790-1795.

Cremer, T. and Cremer, C. (2001). Chromosome territories, nuclear architecture and generegulation in mammalian cells. Nat. Rev. Genet. 2, 292-301.

Croft, J. A., Bridger, J. M., Boyle, S., Perry, P., Teague, P. and Bickmore, W. A. (1999).Differences in the localization and morphology of chromosomes in the human nucleus.J. Cell Biol. 145, 1119-1131.

de Lanerolle, P., Johnson, T. and Hofmann, W. A. (2005). Actin and myosin I in thenucleus: what next? Nat. Struct. Mol. Biol. 12, 742-746.

Gilchrist, S., Gilbert, N., Perry, P., Ostlund, C., Worman, H. J. and Bickmore, W. A.(2004). Altered protein dynamics of disease-associated lamin A mutants. BMC Cell Biol.5, 46.

Goldman, R. D., Gruenbaum, Y., Moir, R. D., Shumaker, D. K. and Spann, T. P.(2002). Nuclear lamins: building blocks of nuclear architecture. Genes Dev. 16, 533-547.

Gruenbaum, Y., Goldman, R. D., Meyuhas, R., Mills, E., Margalit, A., Fridkin, A.,Dayani, Y., Prokocimer, M. and Enosh, A. (2003). The nuclear lamina and its functionsin the nucleus. Int. Rev. Cytol. 226, 1-62.

Harborth, J., Elbashir, S. M., Bechert, K., Tuschl, T. and Weber, K. (2001). Identificationof essential genes in cultured mammalian cells using small interfering RNAs. J. CellSci. 114, 4557-4565.

Hozak, P., Sasseville, A. M. J., Raymond, Y. and Cook, P. R. (1995). Lamin proteinsform an internal nucleoskeleton as well as a peripheral lamina in human cells. J. CellSci. 108, 635-644.

Jackson, D. A. (1991). Structure-function relationships in Eukaryotic nuclei. BioEssays13, 1-10.

Jackson, D. A., Hassan, A. B., Errington, R. J. and Cook, P. R. (1993). Visualizationof focal sites of transcription within human nuclei. EMBO J. 12, 1059-1065.

Lammerding, J., Fong, L. G., Ji, J. Y., Reue, K., Stewart, C. L., Young, S. G. and Lee,R. T. (2006). Lamins A and C but not lamin B1 regulate nuclear mechanics. J. Biol.Chem. 281, 25768-25780.

Lamond, A. I. and Spector, D. L. (2003). Nuclear speckles: a model for nuclear organelles.Nat. Rev. Mol. Cell Biol. 4, 605-612.

Malhas, A., Lee, C. F., Sanders, R., Saunders, N. J. and Vaux, D. J. (2007). Defects inlamin B1 expression and processing affect interphase chromosome position and geneexpression. J. Cell Biol. 176, 593-603.

Misteli, T. (2004). Spatial positioning; a new dimension in genome function. Cell 119,153-156.

Moir, R. D., Yoon, M., Khuon, S. and Goldman, R. D. (2000). Nuclear lamins A andB1: different pathways of assembly during nuclear envelope formation in living cells.J. Cell Biol. 151, 1155-1168.

Mounkes, L., Kozlov, S., Burke, B. and Stewart, C. L. (2003). The laminopathies: nuclearstructure meets disease. Curr. Opin. Genet. Dev. 13, 223-230.

Pombo, A., Jackson, D. A., Hollinshead, M., Wang, Z., Roeder, R. G. and Cook, P. R.(1999). Regional specialization in human nuclei: visualization of discrete sites oftranscription by RNA polymerase III. EMBO J. 18, 2241-2253.

Raska, I., Shaw, P. J. and Cmarko, D. (2006). Structure and function of the nucleolusin the spotlight. Curr. Opin. Cell Biol. 18, 325-334.

Robertson, J. D., Orrenius, S. and Zhivotosky, B. (2000). Nuclear events in apoptosis.J. Struct. Biol. 129, 346-358.

Roos, W. P. and Kaina, B. (2006). DNA damage-induced cell death by apoptosis. TrendsMol. Med. 12, 440-450.

Spann, T. P., Goldman, A. E., Wang, C., Huang, S. and Goldman, R. D. (2002).Alteration of nuclear lamin organization inhibits RNA polymerase II-dependenttranscription. J. Cell Biol. 156, 603-608.

Spector, D. L. (1993). Macromolecular domains within the cell nucleus. Annu. Rev. CellBiol. 9, 265-315.

Spilianakis, C. G. and Flavell, R. A. (2006). Managing associations between differentchromosomes. Science 312, 207-208.

Sullivan, T., Escalante-Alcalde, D., Bhatt, H., Anver, M., Bhat, N., Nagashima, K.,Stewart, C. L. and Burke, B. (1999). Loss of A-type lamin expression compromisesnuclear envelope integrity leading to muscular dystrophy. J. Cell Biol. 147, 913-919.

Vergnes, L., Peterfy, M., Bergo, M. O., Young, S. G. and Reue, K. (2004). Lamin B1is required for mouse development and nuclear integrity. Proc. Natl. Acad. Sci. USA101, 10428-10433.

Worman, H. J. and Courvalin, J. C. (2005). Nuclear envelope, nuclear lamina, andinherited disease. Int. Rev. Cytol. 246, 231-279.

Journal of Cell Science 121 (7)1024

Jour

nal o

f Cel

l Sci

ence