+ All Categories
Home > Documents > A new mechanism controlling kinetochore–microtubule...

A new mechanism controlling kinetochore–microtubule...

Date post: 21-Sep-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
16
A new mechanism controlling kinetochore–microtubule interactions revealed by comparison of two dynein-targeting components: SPDL-1 and the Rod/Zwilch/Zw10 complex Reto Gassmann, 1 Anthony Essex, 1,9 Jia-Sheng Hu, 1,5,9 Paul S. Maddox, 1,6 Fumio Motegi, 2,7 Asako Sugimoto, 2 Sean M. O’Rourke, 3 Bruce Bowerman, 3 Ian McLeod, 4 John R. Yates III, 4 Karen Oegema, 1 Iain M. Cheeseman, 1,8 and Arshad Desai 1,10 1 Ludwig Institute for Cancer Research/Dept of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA; 2 Laboratory for Developmental Genomics, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; 3 Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403, USA; 4 Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA Chromosome segregation requires stable bipolar attachments of spindle microtubules to kinetochores. The dynein/dynactin motor complex localizes transiently to kinetochores and is implicated in chromosome segregation, but its role remains poorly understood. Here, we use the Caenorhabditis elegans embryo to investigate the function of kinetochore dynein by analyzing the Rod/Zwilch/Zw10 (RZZ) complex and the associated coiled-coil protein SPDL-1. Both components are essential for Mad2 targeting to kinetochores and spindle checkpoint activation. RZZ complex inhibition, which abolishes both SPDL-1 and dynein/dynactin targeting to kinetochores, slows but does not prevent the formation of load-bearing kinetochore–microtubule attachments and reduces the fidelity of chromosome segregation. Surprisingly, inhibition of SPDL-1, which abolishes dynein/dynactin targeting to kinetochores without perturbing RZZ complex localization, prevents the formation of load-bearing attachments during most of prometaphase and results in extensive chromosome missegregation. Coinhibition of SPDL-1 along with the RZZ complex reduces the phenotypic severity to that observed following RZZ complex inhibition alone. We propose that the RZZ complex can inhibit the formation of load-bearing attachments and that this activity of the RZZ complex is normally controlled by dynein/dynactin localized via SPDL-1. This mechanism could coordinate the hand-off from initial weak dynein-mediated lateral attachments, which help orient kinetochores and enhance their ability to capture microtubules, to strong end-coupled attachments that drive chromosome segregation. [Keywords: Centromere; aneuploidy; mitosis; kinetochore; microtubule; spindle; chromosome] Supplemental material is available at http://www.genesdev.org. Received April 22, 2008; revised version accepted July 18, 2008. In higher eukaryotes, kinetochores are built on the cen- tromere region of chromosomes to connect to the micro- tubules of the nascent mitotic spindle after nuclear en- velope breakdown (NEBD). To avoid chromosome loss, kinetochores must be efficient at capturing microtu- bules emanating from the two spindle poles and at con- verting initial transient contacts into stable end-coupled attachments capable of resisting the forces that drive chromosome alignment (Nicklas 1988). A safeguard is provided by the mitotic spindle checkpoint, which de- lays cell cycle progression by producing a diffusible in- hibitor at kinetochores that have not yet captured mi- crotubules (Musacchio and Salmon 2007). Stable end-on attachments shut off production of the inhibitory signal, allowing the cell to exit mitosis. The core microtubule attachment site at the kineto- chores is formed by a set of conserved interacting pro- teins, collectively named the KMN network after its Present addresses: 5 Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, Irvine, CA 92697, USA; 6 Institute for Research in Immunology and Cancer, Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec H3C 3J7, Canada; 7 Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA; 8 White- head Institute for Biomedical Research, Cambridge, MA 02142, USA. 9 These authors contributed equally to this work. 10 Corresponding author. E-MAIL [email protected]; FAX (858)-534-7750. Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1687508. GENES & DEVELOPMENT 22:2385–2399 © 2008 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/08; www.genesdev.org 2385 Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.org Downloaded from
Transcript
Page 1: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

A new mechanism controllingkinetochore–microtubule interactionsrevealed by comparison of twodynein-targeting components: SPDL-1and the Rod/Zwilch/Zw10 complexReto Gassmann,1 Anthony Essex,1,9 Jia-Sheng Hu,1,5,9 Paul S. Maddox,1,6 Fumio Motegi,2,7

Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John R. Yates III,4

Karen Oegema,1 Iain M. Cheeseman,1,8 and Arshad Desai1,10

1Ludwig Institute for Cancer Research/Dept of Cellular and Molecular Medicine, University of California at San Diego,La Jolla, California 92093, USA; 2Laboratory for Developmental Genomics, RIKEN Center for Developmental Biology,Kobe 650-0047, Japan; 3Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403, USA; 4Department ofCell Biology, The Scripps Research Institute, La Jolla, California 92037, USA

Chromosome segregation requires stable bipolar attachments of spindle microtubules to kinetochores. Thedynein/dynactin motor complex localizes transiently to kinetochores and is implicated in chromosomesegregation, but its role remains poorly understood. Here, we use the Caenorhabditis elegans embryo toinvestigate the function of kinetochore dynein by analyzing the Rod/Zwilch/Zw10 (RZZ) complex and theassociated coiled-coil protein SPDL-1. Both components are essential for Mad2 targeting to kinetochores andspindle checkpoint activation. RZZ complex inhibition, which abolishes both SPDL-1 and dynein/dynactintargeting to kinetochores, slows but does not prevent the formation of load-bearing kinetochore–microtubuleattachments and reduces the fidelity of chromosome segregation. Surprisingly, inhibition of SPDL-1, whichabolishes dynein/dynactin targeting to kinetochores without perturbing RZZ complex localization, preventsthe formation of load-bearing attachments during most of prometaphase and results in extensive chromosomemissegregation. Coinhibition of SPDL-1 along with the RZZ complex reduces the phenotypic severity to thatobserved following RZZ complex inhibition alone. We propose that the RZZ complex can inhibit theformation of load-bearing attachments and that this activity of the RZZ complex is normally controlled bydynein/dynactin localized via SPDL-1. This mechanism could coordinate the hand-off from initial weakdynein-mediated lateral attachments, which help orient kinetochores and enhance their ability to capturemicrotubules, to strong end-coupled attachments that drive chromosome segregation.

[Keywords: Centromere; aneuploidy; mitosis; kinetochore; microtubule; spindle; chromosome]

Supplemental material is available at http://www.genesdev.org.

Received April 22, 2008; revised version accepted July 18, 2008.

In higher eukaryotes, kinetochores are built on the cen-tromere region of chromosomes to connect to the micro-tubules of the nascent mitotic spindle after nuclear en-velope breakdown (NEBD). To avoid chromosome loss,

kinetochores must be efficient at capturing microtu-bules emanating from the two spindle poles and at con-verting initial transient contacts into stable end-coupledattachments capable of resisting the forces that drivechromosome alignment (Nicklas 1988). A safeguard isprovided by the mitotic spindle checkpoint, which de-lays cell cycle progression by producing a diffusible in-hibitor at kinetochores that have not yet captured mi-crotubules (Musacchio and Salmon 2007). Stable end-onattachments shut off production of the inhibitory signal,allowing the cell to exit mitosis.

The core microtubule attachment site at the kineto-chores is formed by a set of conserved interacting pro-teins, collectively named the KMN network after its

Present addresses: 5Department of Pathology and Laboratory Medicine,University of California Irvine School of Medicine, Irvine, CA 92697,USA; 6Institute for Research in Immunology and Cancer, Department ofPathology and Cell Biology, University of Montreal, Montreal, QuebecH3C 3J7, Canada; 7Department of Molecular Biology and Genetics,Johns Hopkins School of Medicine, Baltimore, MD 21218, USA; 8White-head Institute for Biomedical Research, Cambridge, MA 02142, USA.9These authors contributed equally to this work.10Corresponding author.E-MAIL [email protected]; FAX (858)-534-7750.Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1687508.

GENES & DEVELOPMENT 22:2385–2399 © 2008 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/08; www.genesdev.org 2385

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 2: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

constituent components KNL-1, the Mis12 complex, andthe Ndc80 complex (Cheeseman et al. 2004). The net-work contains two microtubule-binding sites, one inKNL-1 and the other in the Ndc80 complex (Cheesemanet al. 2006; Wei et al. 2007). In eukaryotes ranging fromyeast to human cells, compromising Ndc80 complexfunction in vivo leads to severe chromosome alignmentdefects correlated with an inability of kinetochores to formstable bipolar attachments (Kline-Smith et al. 2005).

Additional kinetochore–microtubule interactions aremediated by the microtubule minus-end-directed motorcytoplasmic dynein and its cofactor dynactin. Becausedynein/dynactin has multiple functions in the cell, in-sight into its kinetochore roles has primarily come fromstudies on the conserved Rod/Zwilch/Zw10 (RZZ) com-plex (Smith et al. 1985; Karess and Glover 1989; Wil-liams and Goldberg 1994; Scaerou et al. 1999, 2001;Williams et al. 2003), which is essential for kinetochorerecruitment of dynein/dynactin (Starr et al. 1998). Inhi-bitions of RZZ subunits (Savoian et al. 2000; Li et al.2007; Yang et al. 2007) and direct disruption of dynein/dynactin (Vorozhko et al. 2008) have shown that the mi-nus-end-directed motility of kinetochore dynein contrib-utes to transient poleward movement of chromosomesin early prometaphase (Rieder and Alexander 1990). In-hibition of dynein/dynactin also affects the microtubule-based poleward transport of checkpoint proteins andRZZ subunits, which is thought to constitute an impor-tant mechanism for silencing the spindle checkpoint(Howell et al. 2001; Wojcik et al. 2001).

Despite significant work over the past decade, the rel-evance of kinetochore dynein/dynactin for the process ofchromosome alignment remains unclear. When dynein/dynactin is inhibited following bipolar spindle assembly,metaphase plate formation occurs normally (Howell etal. 2001; Vorozhko et al. 2008). Similarly, Drosophilamelanogaster null mutations in the rod and zw10 geneswere reported as having no obvious phenotype prior toanaphase in mitotic cells (Williams et al. 1992; Williamsand Goldberg 1994). In contrast, recent work in mamma-lian cells showed significant delays in chromosomealignment after depletion of Zw10 (Li et al. 2007; Yang etal. 2007). The dissection of RZZ complex function inchromosome alignment is complicated by its role inspindle checkpoint signaling. Analysis in D. melanogas-ter embryos, human cells, and Xenopus extracts demon-strated that the RZZ complex is essential for spindlecheckpoint function (Basto et al. 2000; Chan et al. 2000;Kops et al. 2005) and for the localization to unattachedkinetochores of two essential spindle checkpoint com-ponents, Mad1 and Mad2 (Buffin et al. 2005; Kops et al.2005).

The detection of a two-hybrid interaction betweenZw10 and the dynactin subunit dynamitin suggestedthat the RZZ complex is directly involved in recruitingdynein/dynactin (Starr et al. 1998). A recent study in D.melanogaster identified Spindly, a component actingdownstream from the RZZ complex, which is requiredfor targeting dynein, but not dynactin, to kinetochores(Griffis et al. 2007). NudE and NudEL, two proteins that

associate with dynein, have also been implicated indynein targeting to kinetochores (Stehman et al. 2007;Vergnolle and Taylor 2007).

We developed the early Caenorhabditis elegans em-bryo as a system to identify proteins that play importantroles in chromosome segregation and characterize theirmechanism of action (Oegema et al. 2001; Desai et al.2003; Cheeseman et al. 2004). Here, we use this systemto study the function of the Rod/Zwilch/Zw10 (RZZ)complex and Spindly (SPDL-1), components of the outerkinetochore that are essential for spindle checkpointfunction and constitute a module that targets thedynein/dynactin motor to the kinetochore during chro-mosome alignment. A comparative analysis of SPDL-1and RZZ complex function revealed that, despite theirequivalent requirement for dynein/dynactin recruit-ment, SPDL-1 inhibition results in a significantly moresevere defect in chromosome segregation, which, untiljust prior to anaphase onset, closely mimics the lack ofstable end-coupled attachments. This defect can bequantitatively reduced to match that of inhibiting theRZZ complex alone by coinhibiting SPDL-1 and the RZZcomplex. Thus, by uncoupling kinetochore localizationof the RZZ complex from that of dynein/dynactin, weuncovered a regulatory relationship between the RZZcomplex and the formation of stable end-coupled attach-ments. We discuss the implications of these findings fordynein/dynactin function at the kinetochore.

Results

The C. elegans Spindly homolog C06A8.5/SPDL-1is required for chromosome segregation

All C. elegans proteins essential for chromosome segre-gation are required for embryonic viability. Embryos in-dividually depleted of each of the ∼2000 gene productsrequired for embryonic viability have been filmed usingdifferential interference contrast (DIC) microscopy toidentify genes whose inhibition results in the presence ofextra nuclei (karyomeres) due to chromosome missegre-gation (Sonnichsen et al. 2005). However, since misseg-regation does not always lead to the formation of karyo-meres and DIC does not directly visualize chromosomes,it is likely that chromosome segregation genes have beenmissed by this approach. To test this idea, we used RNAito target 50 genes of unknown function required for em-bryonic viability but annotated as having no defects byDIC analysis in a strain coexpressing GFP:histone H2Band GFP:�-tubulin to visualize chromosomes andspindle poles, respectively (Oegema et al. 2001). Thisscreen identified one previously uncharacterized gene,C06A8.5, whose depletion resulted in severe chromo-some missegregation. C06A8.5 encodes a 479-amino-acid protein that contains five predicted coiled-coil do-mains in its N-terminal 360 residues. Sequence searchesidentified potential homologs throughout the animalkingdom, including the previously characterized D. mel-anogaster Spindly (Griffis et al. 2007). AlthoughC06A8.5 shows low sequence identity with the other

Gassmann et al.

2386 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 3: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

proteins, it shares a highly conserved motif located neara break in the coiled-coils (Fig. 1C; Supplemental Fig. 1).As our functional analysis supports the idea thatC06A8.5 is a Spindly homolog, we named the C. elegansprotein SPDL-1.

Embryos depleted of SPDL-1 exhibited defective chro-mosome alignment, premature spindle pole separation,and significant chromatin bridges in anaphase (Fig. 1A;

Supplemental Movie 1). The onset of both sister chro-matid separation and cytokinesis occurred with normaltiming in spdl-1(RNAi) embryos, indicating that cellcycle progression was unaffected (Fig. 1B).

The chromosome missegregation observed in spdl-1(RNAi) embryos could be the result of compromisedmitotic chromosome structure, an aberrant mitoticspindle, or a defect in kinetochore function. To distin-

Figure 1. SPDL-1 is a transient kineto-chore component essential for chromo-some segregation. (A) Selected framesfrom a live-imaging sequence of the firstdivision in unperturbed and spdl-1(RNAi)embryos expressing GFP:histone H2Band GFP:�-tubulin to simultaneously vi-sualize chromosomes (arrow) and spindlepoles (arrowheads), respectively (see alsoSupplemental Movie 1). Images are time-aligned relative to NEBD (0 sec). Bar, 5 µm.(B) Timing of anaphase onset and cytoki-nesis onset in unperturbed and spdl-1(RNAi) embryos. Anaphase onset was de-fined as the first visible sister chromatidseparation (GFP:histone H2B) and cytoki-nesis onset by the first visible ingressionof the cleavage furrow in DIC images ac-quired in parallel. Values represent theS.E.M with a 95% confidence interval. (C)Primary sequence features of SPDL-1 andrelated proteins. The highly conservedmotif that defines this conserved coiled-coil protein family is depicted (see alsoSupplemental Fig. 1). (D) Chromosomecondensation, sister centromere resolu-tion, and the separation of sister chroma-tids at anaphase onset are normal in spdl-1(RNAi) embryos. Selected frames of alive-imaging sequence are shown (see alsoSupplemental Movie 2). Kinetochores aremarked by GFP:Spc24KBP-4, a subunit ofthe NDC-80 complex. Arrows highlightseparating sister kinetochores at anaphaseonset (0 sec) in spdl-1(RNAi) embryos. Bar,5 µm. (E) Mitotic spindle morphology incontrol and spdl-1(RNAi) embryos fixedand stained with a fluorescently labeledantibody against �-tubulin (see alsoSupplemental Movie 3). Bar, 5 µm. (F) Im-munoblotting with an affinity-purifiedpolyclonal antibody raised against SPDL-1detects purified recombinant (rec.) SPDL-1and a protein of equal size in wild-type(N2) worms, which is depleted >95% byRNAi. The relative amount of worm ex-tract loaded is indicated above each lane.A cross-reacting protein band (*) serves asthe loading control. (G) Immunofluores-cence image of a one-cell embryo at pro-

metaphase immunostained for SPDL-1. Bar, 2 µm. (H) Snapshot of a one-cell embryo in prometaphase expressing GFP:SPDL-1. Bar, 5µm. (I) SPDL-1 localizes transiently to kinetochores from prometaphase to anaphase onset. Two-cell embryos at different stages areshown costained for CENP-CHCP-4, which is present at kinetochores throughout mitosis, and SPDL-1. The natural difference in cellcycle timing of the AB and P1 cells (with AB entering and exiting mitosis prior to P1, as diagrammed on the right) defines the transientperiod of SPDL-1 kinetochore localization (see also Supplemental Movie 4). Bar, 5 µm.

Dynein function at kinetochores

GENES & DEVELOPMENT 2387

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 4: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

guish between these possibilities, we imaged a wormstrain expressing GFP:Spc24KBP-4, a subunit of the outerkinetochore NDC-80 complex (Cheeseman et al. 2004).In spdl-1(RNAi) embryos, GFP:Spc24KBP-4 localized nor-mally to paired diffuse kinetochores that maintained arigid parallel conformation during prometaphase chro-mosome movements, demonstrating that centromereresolution and chromosome condensation were unaf-fected (Fig. 1D; Supplemental Movie 2). Sister kineto-chores remained paired until the onset of anaphase,when individual chromatids separated from each other(Fig. 1D, arrows), indicating proper regulation of sisterchromatid cohesion. All microtubule-dependent eventsin the early embryo (pronuclear migration, rotation ofthe centrosome–pronuclear complex, spindle assembly,and asymmetric spindle positioning) were normal inspdl-1(RNAi) embryos. In particular, fixed and liveanalysis in a worm strain expressing GFP:�-tubulinshowed that mitotic spindle formation in spdl-1(RNAi)embryos was not perturbed (Fig. 1E; Supplemental Movie3). We conclude that the severe chromosome segregationdefect in embryos depleted of SPDL-1 does not resultfrom problems with either the microtubule cytoskele-ton, mitotic spindle formation, or chromosome struc-ture.

Immunoblotting using affinity-purified antibodiesconfirmed that our RNAi conditions resulted in pen-

etrant depletion of SPDL-1 (Fig. 1F). Immunostaining re-vealed that SPDL-1 is recruited to kinetochores at NEBDand localizes there until the metaphase–anaphase tran-sition, after which it is no longer detected (Fig. 1G,I).Imaging of a worm strain expressing GFP:SPDL-1 con-firmed this transient localization pattern and also re-vealed a weak spindle pole localization (Fig. 1H; Supple-mental Movie 4). We conclude that SPDL-1 is a tran-siently kinetochore-localized protein that plays anessential role in chromosome segregation.

SPDL-1 is recruited to kinetochores by the RZZcomplex

To understand SPDL-1 function at kinetochores, we firstsought to determine if it interacts with other knownkinetochore components. We immunoprecipitated sev-eral inner and outer kinetochore proteins (CENP-AHCP-3,CENP-CHCP-4, MCAKKLP-7, KNL-1, BUB-1, ZwilchZWL-1,NDC-80, HCP-1, CLASPCLS-2) and probed the precipi-tates for SPDL-1. Only affinity-purified antibodies toZwilchZWL-1 (Fig. 2C) coprecipitated a detectableamount of SPDL-1 (Fig. 2A). Mass spectrometric analysisof the ZwilchZWL-1 immunoprecipitate (Fig. 2B) con-firmed the presence of SPDL-1 and identified the twoother subunits of the RZZ complex, ROD-1 andZw10CZW-1. In contrast, a more stringent tandem puri-

Figure 2. SPDL-1 is recruited to the ki-netochore by the RZZ complex. (A)SPDL-1 coimmunoprecipitates withZwilchZWL-1. Worm extracts were de-pleted of ZwilchZWL-1 using an affinity-purified polyclonal antibody, and the re-sulting supernatant (S) and pellet (P) wasanalyzed by immunoblot. Loading of pel-let is 20× relative to supernatant. An an-tibody against GFP was used in thecontrol immunoprecipitation experi-ment. (B) SPDL-1 associates with theRZZ complex but is not a core subunit.A one-step immunoprecipitation ofZwilchZWL-1 and a stringent two-stepisolation of GFPLAP-tagged ZwilchZWL-1

were visualized on a silver-stained geland analyzed by mass spectrometry asshown on the right. (C) Immunoblottingwith the anti-ZwilchZWL-1 antibody de-tects a 70-kDa band, which is depleted>95% following RNAi. A cross-reactingprotein band (*) serves as the loadingcontrol. (D) Immunofluorescence imagesof early embryos stained for SPDL-1 afterdepletion of ZwilchZWL-1 or ROD-1.Bars, 5 µm. (E) Depletion of SPDL-1 af-fects neither kinetochore targeting ofRZZ subunits nor their rapid disappear-ance from kinetochores at anaphase on-set. Selected frames from time-lapse se-quences of embryos expressing GFP:ZwilchZWL-1, GFP:Zw10CZW-1, and GFP:Spc24KBP-4 are shown (see also Supplemental Movies 5,6). Time is relative to the onset of sister chromatid separation. Bar, 5 µm.

Gassmann et al.

2388 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 5: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

fication of ZwilchZWL-1 using the localization and affin-ity purification (LAP) tag (Cheeseman et al. 2004) iso-lated only the three RZZ subunits, which could bereadily visualized on a silver-stained gel (Fig. 2B), but notSPDL-1, suggesting that SPDL-1 is peripherally associ-ated rather than a stable core subunit of the RZZ com-plex. Consistent with this idea, gel filtration experi-ments revealed that endogenous SPDL-1 exhibited thesame fractionation profile as recombinant SPDL-1, andthat this fractionation behavior was clearly distinct fromthat of endogenous ZwilchZWL-1 (data not shown).

We next analyzed the localization dependencies be-tween SPDL-1 and the RZZ complex. Immunofluores-cence and live imaging of GFPLAP fusions toZwilchZWL-1 and Zw10CZW-1 showed that these twoRZZ subunits localize transiently to kinetochores in afashion essentially identical to SPDL-1 (Fig. 2D,E;Supplemental Movies 5, 6). ROD-1 depletion abolishedkinetochore localization of both Zw10CZW-1 andZwilchZWL-1, suggesting that the three RZZ subunits arelikely interdependent for kinetochore targeting (Fig. 2D;data not shown). Depletion of ZwilchZWL-1 or ROD-1abolished SPDL-1 targeting to kinetochores (Fig. 2D),whereas SPDL-1 depletion did not alter the kinetics ofkinetochore localization for either RZZ subunit (Fig. 2E;Supplemental Movies 5, 6). Specifically, rapid disappear-ance of ZwilchZWL-1 and ROD-1 from kinetochores inearly anaphase was observed in both control and spdl-1(RNAi) embryos. This loss occurred prior to mitotickinetochore disassembly, monitored using the NDC-80complex subunit Spc24KBP-4, suggesting the existence ofa regulatory step controlling RZZ complex removal fromkinetochores that is not affected by SPDL-1 depletion.We conclude that SPDL-1 is recruited to kinetochores bythe RZZ complex and that both the localization and cellcycle progression-dependent loss of the RZZ complexfrom kinetochores are independent of SPDL-1.

SPDL-1 and the RZZ complex are dispensablefor building the core kinetochore microtubuleattachment site

Next we positioned SPDL-1 and the RZZ complexwithin the established hierarchy for kinetochore assem-bly. SPDL-1 and RZZ complex localization was depen-dent on KNL-1 (Fig. 3A), which is required for the local-ization of multiple outer kinetochore proteins in C. el-egans (Desai et al. 2003). Depletion of the CENP-F-likeproteins HCP-1 and HCP-2 had no effect on SPDL-1 lo-calization. An intermediate effect on SPDL-1 localiza-tion was observed following depletion of NDC-80 orBUB-1; the effect of BUB-1 depletion was consistentlymore severe than that of NDC-80 depletion. RZZ com-plex targeting was not affected by any of these depletions(Fig. 3A; A. Essex and A. Desai, unpubl.).

Depletion of SPDL-1 and RZZ subunits had no effecton the localization of KNL-1, KNL-2, KNL-3, MIS-12,NDC-80, the NDC-80 complex subunit Spc25KBP-3,BUB-1, HCP-1, or CLASPCLS-2 (Fig. 3B; data not shown).We conclude that outer kinetochore assembly, including

formation of the core microtuble attachment site consti-tuted by the KMN network, does not require either theRZZ complex or SPDL-1 (Fig. 3C).

SPDL-1, like the RZZ complex, is requiredfor a functional spindle checkpoint and Mad2MDF-2

recruitment to unattached kinetochores

Having established that SPDL-1 functions in close prox-imity to the RZZ complex, we sought to test if any of theroles ascribed to the RZZ complex require SPDL-1. Workin D. melanogaster and vertebrates has shown that theRZZ complex is required for a functional spindle check-point and for localization of the checkpoint proteinMad2 to kinetochores. To probe spindle checkpoint ac-tivation in the early C. elegans embryo, we used an assaybased on controlled formation of monopolar spindles inthe second embryonic division following inhibition ofcentriole duplication (A. Essex and A. Desai, in prep.).These monopolar spindles elicit a checkpoint-mediatedcell cycle delay (Fig. 4B). Depletion of SPDL-1 in cellswith monopolar spindles abrogated the delay, demon-strating that the spindle checkpoint requires SPDL-1(Fig. 4B). The same result was observed for depletions ofthe RZZ subunit ROD-1. The delay was correlated withtransient enrichment of GFP:Mad2MDF-2 on the unat-tached kinetochores that are distal to the monopole (Fig.4C; Supplemental Movie 7). Depletion of SPDL-1 orROD-1 abrogated GFP:Mad2MDF-2 localization to kineto-chores of monopolar spindles. We conclude that SPDL-1,like the RZZ complex, is required for spindle check-point activation and kinetochore recruitment ofGFP:Mad2MDF-2.

SPDL-1 is required for the recruitment of dyneinand dynactin to unattached kinetochores

Dynein/dynactin recruitment to kinetochores has beenhypothesized to occur via the dynactin subunit p50/dy-namitin, a two-hybrid interactor of Zw10 (Starr et al.1998). D. melanogaster Spindly (DmSpindly) was re-ported to be required for dynein but not dynactin target-ing to kinetochores, suggesting that dynactin-Zw10 andDmSpindly make independent contributions to dyneinlocalization (Griffis et al. 2007). To test if this is the casein C. elegans, we generated worm strains stably coex-pressing mCherry:histone H2B and GFP:fusions of cyto-plasmic dynein heavy chainDHC-1 or dynamitinDNC-2.Both fusion proteins localized diffusely to the spindleand the spindle poles in mitosis and significant enrich-ment at kinetochores over the spindle signal was notevident in unperturbed embryos (data not shown). How-ever, in cells with monopolar spindles, GFP:dyneinheavy chainDHC-1 and GFP:dynamitinDNC-2 becameprominently enriched at kinetochores (Fig. 5A; Supple-mental Movie 8). Both fusion proteins were excludedfrom the nucleus in interphase and localized to thenuclear periphery opposite the single spindle pole priorto NEBD. After NEBD, both proteins accumulated at ki-

Dynein function at kinetochores

GENES & DEVELOPMENT 2389

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 6: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

netochores. This behavior was distinct from that ofGFP:KNL-2, a centromeric chromatin protein (Maddoxet al. 2007), whose levels at kinetochores did not appre-ciably change throughout monopolar mitosis (Fig. 5A).GFP:KNL-2 was also present on all sister kinetochoresregardless of their orientation relative to the spindlepole. In contrast, the accumulation of both GFP:dyneinheavy chainDHC-1 and GFP:dynamitinDNC-2 was restrictedto kinetochores that were located on the distal, unattachedside of the monopolar spindle. These results indicate thatboth dynein and dynactin accumulate at unattached ki-netochores in C. elegans, as is the case in vertebrates.

We next investigated the role of SPDL-1 in the accu-mulation of dynein and dynactin on kinetochores of mo-nopolar spindles. We found that both GFP:dynein heavychainDHC-1 and GFP:dynamitinDNC-2 failed to localize tokinetochores of monopolar spindles in spdl-1(RNAi) em-

bryos (Fig. 5B; Supplemental Movies 9, 10). A similarresult was observed following RZZ complex inhibition.In contrast, depletion of NDC-80 did not prevent kineto-chore localization of either GFP:dynein heavy chainDHC-1

or GFP:dynamitinDNC-2. Since both SPDL-1 and the RZZcomplex are required for the spindle checkpoint-medi-ated delay elicited by monopolar spindles, we usedMad2MDF-2 depletion to test whether the lack of dynein/dynactin localization was caused by accelerated cellcycle progression. Although their gradual accumulationwas cut short by premature mitotic exit, both GFP:dynein heavy chainDHC-1 and GFP:dynamitinDNC-2 werevisible at kinetochores in Mad2MDF-2-depleted cells withmonopolar spindles (Fig. 5C; Supplemental Movies 9,10). Of note, none of the perturbations affected the lo-calization of GFP:dynein heavy chainDHC-1 or GFP:dynamitinDNC-2 to the nuclear periphery. We conclude

Figure 3. SPDL-1 and the RZZ complexare dispensable for the formation of thecore kinetochore microtubule attachmentsite. (A) Consequences of outer kinetochorecomponent depletion on the localization ofSPDL-1 and ZwilchZWL-1, assayed by im-munofluorescence. Bars, 5 µm. (B) Normallocalization of outer kinetochore compo-nents after depletion of SPDL-1 andZwilchZWL-1, assayed by immunofluores-cence or live imaging of previously charac-terized GFP-fusions (Cheeseman et al.2004, 2005; Maddox et al. 2007). Bars, 5 µm.(C) Summary of the dependency analysisfor kinetochore targeting of SPDL-1 and theRZZ complex. For each depletion-localiza-tion experiment, between five and 10 one-cell or two-cell embryos were examined.

Gassmann et al.

2390 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 7: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

that kinetochore localization of both dynein and dynac-tin is dependent on SPDL-1.

Depletion of SPDL-1 results in a more severe defectin kinetochore–microtubule attachments thandepletion of RZZ complex subunits

SPDL-1 localizes downstream from the RZZ complexand is required for all RZZ complex functions estab-

lished to date: spindle checkpoint activation, kineto-chore recruitment of Mad2, and kinetochore recruitmentof dynein/dynactin. These findings predict that chromo-some segregation defects in spdl-1(RNAi) embryosshould be of similar or reduced severity compared withthose observed in embryos depleted of RZZ subunits. Wefirst tested whether depletion of the three RZZ subunitsresulted in embryonic lethality, as is the case for deple-tion of SPDL-1. This analysis confirmed the results offunctional genomic studies (Sonnichsen et al. 2005) thatthe three RZZ subunits are not functionally equivalent.Depletion of Zw10CZW1 causes penetrant sterilityof the injected worm, whereas depletion of ROD-1 orZwilchZWL-1 results in embryonic lethality of its proge-ny (A. Essex and A. Desai, in prep.). This difference isexplained by the requirement of Zw10CZW1, but notROD-1 or ZwilchZWL-1, for membrane trafficking in thegonad (Anjon Audhya, pers. comm.); defects in traffick-ing pathways prevent oocyte formation and cause steril-ity in C. elegans. Consequently, we focused on ROD-1and ZwilchZWL-1 to specifically analyze RZZ complexfunction at kinetochores.

We compared the consequences of SPDL-1 depletionwith those of ROD-1 or ZwilchZWL-1 depletions in astrain coexpressing GFP:histone H2B, which allowed vi-sual inspection of chromosome alignment and separa-tion, and GFP:� tubulin, which facilitated spindle poletracking (Fig. 6A). The latter assay is particularly usefulin the one-cell C. elegans embryo, where kinetochore–spindle attachments counteract cortical forces pullingon astral microtubules anchored at the spindle poles;premature pole separation following perturbation of ki-netochore-localized proteins is diagnostic of impairmentin the formation of load-bearing kinetochore–microtu-bule attachments, and specific pole separation profileshave proven important in categorizing different types ofdefects (Oegema et al. 2001; Desai et al. 2003; Cheese-man et al. 2004).

Surprisingly, we observed that chromosome segrega-tion and kinetochore–spindle microtubule interactiondefects were significantly worse in SPDL-1-depleted em-bryos than in embryos depleted of RZZ subunits. In spdl-1(RNAi) embryos, which never congressed their chromo-somes to a compact metaphase plate (Fig. 6B), the poleseparation profile was identical to that of knl-3(RNAi)embryos in which outer kinetochore assembly was pre-vented (Cheeseman et al. 2004), except for a short period(∼40 sec) prior to sister chromatid separation (Fig. 6E).Although pole separation slowed during this period, sug-gesting engagement of spindle microtubules by kineto-chores, spindles were significantly longer at the time ofsister chromatid separation in spdl-1(RNAi) embryos(20 ± 0.5 µm) compared with controls (16.2 ± 0.5 µm). Bycomparison, the chromosome segregation and prematurepole separation defects in rod-1(RNAi) or zwl-1 (RNAi)embryos were markedly less severe (Fig. 6B,F; Supple-mental Movie 11). Chromosomes were able to congressand form a metaphase plate, and sister chromatid sepa-ration appeared successful in the majority of embryos. Asmall amount of lagging anaphase chromatin was con-

Figure 4. SPDL-1 is required for a functional spindle check-point and kinetochore localization of Mad2MDF-2. (A) Perturba-tion to generate monopolar spindles in the second division andtrigger spindle checkpoint activation in C. elegans embryos.ZYG-1 is a kinase required for centriole duplication (O’Connellet al. 2001). In zyg-1(RNAi) embryos, the first division is nor-mal, because two intact centrioles are contributed by spermthat is not affected by RNAi. These centrioles are unable toduplicate, however, resulting in a monopolar spindle in the sub-sequent division. (B) Average time from NEBD to chromosomedecondensation in the P1 cell of a worm strain expressingGFP:histone H2B. ZYG-1 single depletion results in a signifi-cant delay that depends on Mad2MDF-2, SPDL-1, and ROD-1.Error bars represent the SEM with a 95% confidence interval. Asimilar result is observed in the AB cell (data not shown).(C) Stills from a time-lapse sequence of the AB cell monopolardivision in a worm strain coexpressing GFP:Mad2MDF-2 andmCherry:histone H2B. In ZYG-1 single depletions,GFP:Mad2MDF-2 accumulates on kinetochores that are distal tothe pole (arrow). Codepletion of ZYG-1 with SPDL-1 or ROD-1prevents kinetochore accumulation of GFP:Mad2MDF-2 (see alsoSupplemental Movie 7). Bar, 5 µm.

Dynein function at kinetochores

GENES & DEVELOPMENT 2391

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 8: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

sistently observed in ∼30% of first divisions in RZZ sub-unit depletions (Fig. 6D), whereas SPDL-1 depletion re-sulted in massive chromatin bridges in every first divi-sion examined. Pole tracking analysis also revealed asignificantly less severe defect in rod-1 or zwl-1(RNAi)embryos compared with spdl-1(RNAi) embryos (Fig. 6F).In RZZ subunit depletions, initial premature pole sepa-ration was observed indicating a defect in establishingload-bearing attachments. However, ∼100 sec prior tosister chromatid separation, kinetochores engagedspindle microtubules and spindle length at the time ofsister chromatid separation [zwl-1(RNAi), 16 ± 0.3 µm;rod-1(RNAi), 15.6 ± 0.2 µm] was indistinguishable fromcontrols (16.2 ± 0.5 µm).

Both quantitative immunoblotting (Figs. 1F, 2C) andimmunofluorescence (Figs. 2D, 3B) indicate that the dif-ference in phenotypic severity described above is un-likely due to reduced depletion efficiency of RZZ sub-units relative to SPDL-1. The identical qualitative and

quantitative phenotypes of rod-1(RNAi) and zwl-1(RNAi) also argue against this trivial explanation. Tofurther establish that the weaker RZZ inhibition pheno-type is not due to partial depletion of the targeted sub-units, we codepleted ROD-1 and ZwilchZWL-1. The ob-served phenotypes in both assays were indistinguishablefrom single depletions of ROD-1 or ZwilchZWL-1 (Fig. 6C;Supplemental Fig. 2; Supplemental Movie 12).

Thus, RZZ complex inhibition slows but does not pre-vent formation of load-bearing kinetochore–microtubuleattachments and leads to an increase in anaphase laggingchromatin, which is indicative of incorrectly attachedkinetochores. Depletion of SPDL-1 results in signifi-cantly more severe defects both in the formation of load-bearing kinetochore–microtubule attachments and chro-mosome segregation. Based on the pole-tracking analy-sis, the severity of the SPDL-1 inhibition defects closelyresembles loss of core microtubule attachment until justprior to anaphase onset.

Figure 5. Localization of dynein and dy-nactin to kinetochores requires SPDL-1.(A) Unattached kinetochores on second-di-vision monopolar spindles accumulatedynein and dynactin. Strains stably coex-pressing GFP:fusions of either KNL-2, full-length dynein heavy chainDHC-1, or dyna-mitinDNC-2 with mCherry:histone H2Bwere used to monitor kinetochore local-ization (see also Supplemental Movie 8).All images are of the AB cell, and thesingle spindle pole is always to the left.Times are relative to NEBD. Line scans (5pixels wide; normalized relative to maxi-mum intensity in each channel) indicatethe bilaterally symmetric distribution ofKNL-2 relative to mCherry:histone H2B,which contrasts with the asymmetric en-richment of DHC-1 and DNC-2 on thechromosomal face pointing away from thesingle pole. Bars, 5 µm. (B) Kinetochore ac-cumulation of GFP:dynein heavy chain-DHC-1 and GFP:dynamitinDNC-2 requiresSPDL-1 and the RZZ complex but notNDC-80. For brevity, a single frame isshown for each condition (see also Supple-mental Movies 9, 10). Bars, 5 µm. (C) Ab-rogating the spindle checkpoint by deplet-ing Mad2MDF-2 does not prevent recruit-ment of GFP:dynein heavy chainDHC-1

or GFP:dynamitinDNC-2 to unattached ki-netochores. However, accumulation of theGFP:fusion proteins is limited because ofpremature mitotic exit. Bars, 5 µm.

Gassmann et al.

2392 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 9: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

Coinhibition of SPDL-1 with RZZ subunits resultsin the less severe RZZ inhibition phenotype

The greater phenotypic severity of SPDL-1 inhibitionsnoted above may reflect additional nonkinetochore func-tions of SPDL-1 that are not affected by its displacementfrom the kinetochore in RZZ subunit depletions. To testthis possibility, we codepleted ROD-1 or ZwilchZWL-1

with SPDL-1. In all embryos analyzed (n = 32), the result-ing phenotype was indistinguishable from the weakerRZZ subunit depletions (Fig. 6C,G; Supplemental Movie12). To control for reduced efficacy of RNAi in the

double depletions, the SPDL-1 single depletions were re-peated after appropriate dilution with control dsRNA.The reduction in phenotypic severity in the doubledepletions was evident in both the chromosome segre-gation profile (Fig. 6C) and in the quantitative analysis ofspindle pole separation (Fig. 6G). These results are con-sistent with the assembly epistasis at kinetochores,which showed that SPDL-1 depends on the RZZ com-plex for localization (Fig. 2), and exclude the possibilitythat the additional defects observed in spdl-1(RNAi) em-bryos are due to nonkinetochore functions of SPDL-1.We conclude that the severe defects in SPDL-1-depleted

Figure 6. Depletion of SPDL-1 results ina more severe chromosome segrega-tion defect than depletion of ROD-1 orZwilchZWL-1. (A) Cartoon outlining thetwo parameters monitored in a strain ex-pressing GFP:histone H2B and GFP:�-tu-bulin: chromosome dynamics and kineticsof spindle pole separation. (B) Frames fromtime-lapse sequences of the first embry-onic division, highlighting the differencesin chromosome dynamics after depletionof SPDL-1 and the RZZ complex subunitsROD-1 and ZwilchZWL-1 (see also Supple-mental Movie 11). The time point 0 secdenotes the onset of sister chromatid sepa-ration. Bar, 5 µm. (C) Selected frames fromtime-lapse sequences of embryos code-pleted of RZZ subunits and SPDL-1,which significantly reduces the severechromosome missegregation phenotype ofSPDL-1 single depletions to match that ofRZZ subunit single depletions (see alsoSupplemental Movie 12). Bar, 5 µm. (D)Representative image of anaphase withlagging chromatin in a rod-1(RNAi) one-cell embryo. The frequency of one-cell em-bryos with lagging anaphase chromatin isindicated for single and double inhibitionsinvolving RZZ subunits and SPDL-1. Bar,2 µm. (E) Pole separation kinetics in wild-type, spdl-1(RNAi), and knl-3(RNAi) em-bryos. Images were acquired at 10-sec in-tervals, and sequences were time-alignedrelative to NEBD. Pole–pole distances inthe time-aligned sequences were mea-sured, averaged for the indicated number(n) of embryos, and plotted against time.Error bars represent the SEM with a 95%confidence interval. (F) Pole separation ki-netics of the perturbations shown in B. Se-quences were time-aligned relative to theonset of sister chromatid separation(“Anaphase Onset”). Error bars representthe SEM with a 95% confidence interval.(G) Pole separation kinetics of the pertur-bations shown in C, demonstrating thatdouble depletions of SPDL-1 and RZZ

complex subunits result in a pole separation profile that is indistinguishable from RZZ subunit single depletions. For controls, spdl-1dsRNA was diluted equally with dsRNA corresponding to the budding yeast gene CTF13 or the C. elegans gene sas-5, which is notrequired for the first embryonic division (both conditions gave identical results). Error bars represent the SEM with a 95% confidenceinterval.

Dynein function at kinetochores

GENES & DEVELOPMENT 2393

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 10: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

embryos are derived from RZZ complex localized to ki-netochores in the absence of associated SPDL-1 and/ordynein/dynactin.

Codepletion of SPDL-1 or RZZ subunits with NDC-80synergistically recapitulates the “kinetochore-null”phenotype

Two microtubule-binding entities are independently tar-geted to the C. elegans kinetochore via KNL-1: dynein/dynactin (via the RZZ complex and SPDL-1) and theNDC-80 complex, a component of the KMN network(Fig. 3C). In spdl1(RNAi) embryos, just prior to sisterchromatid separation, pole separation slowed down sig-nificantly and severe chromosome missegregation wasobserved along the spindle axis (Figs. 1A, 6B). We hy-pothesized that the eventual slowing of pole separationin spdl-1(RNAi) embryos reflected belated load-bearingattachments made by the NDC-80 complex, while theresidual chromosome movements in ndc-80(RNAi) em-bryos (Supplemental Movie 13) were mediated by kineto-chore dynein/dynactin. Codepletion experiments con-firmed this view (Fig. 7A,B). While embryos singly de-pleted of SPDL-1 and NDC-80 were partially successfulat congressing their chromosomes to the spindle equatorand at segregating sister chromatids in anaphase, doubledepletions resulted in a phenotype identical to that ofknl-3(RNAi) embryos (Supplemental Movie 13), inwhich outer kinetochore assembly is abolished (Cheese-man et al. 2004). A “kinetochore-null”-like phenotypewas also observed in codepletions of NDC-80 withROD-1 (Supplemental Movie 14). We conclude that it isthe absence of the two independently targeted microtu-bule-interacting components, dynein/dynactin and theNDC-80 complex, that accounts for the synergistic de-fect. These results further indicate that the reduction inpole separation just prior to anaphase onset and the mis-segregation of chromosomes observed in SPDL-1-de-pleted embryos are attributable to the action of theNDC-80 complex.

Discussion

Our analysis of the RZZ complex and SPDL-1, kineto-chore-localized components that are sequentially re-quired for dynein/dynactin targeting, gives new insightinto how this minus-end-directed motor complex con-tributes to chromosome segregation. Specifically, the re-sults suggest that kinetochore dynein/dynactin acceler-ates the formation of load-bearing attachments and pro-vides an important fidelity mechanism, which preventsinappropriate attachments in prometaphase and reducesthe missegregation frequency after anaphase onset. Thisfidelity mechanism likely involves negative regulationof load-bearing kinetochore–microtubule attachmentsby the RZZ complex. We speculate below that this nega-tive regulation is modulated by dynein/dynactin to en-sure the orderly conversion of weak dynein/dynactin-mediated lateral attachments to load-bearing end-coupled attachments during prometaphase.

SPDL-1 targets dynein/dynactin to kinetochoresand is required to activate the spindle checkpoint

SPDL-1 targets to the kinetochore immediately down-stream from the RZZ complex and is not involved in theassembly of the core kinetochore–microtubule-bindingsite constituted by the KMN network. Functional analy-sis showed that SPDL-1 is required for the recruitment ofdynein/dynactin and Mad2MDF-2 to unattached kineto-chores. The requirement for Mad2MDF-2 targeting ex-plains why SPDL-1 is essential for spindle checkpointactivation. In contrast, D. melanogaster Spindly wasshown to be essential for the recruitment of dynein, butnot dynactin, to kinetochores, and was found to be dis-pensable for Mad2 accumulation and spindle checkpointactivation. We also did not observe any abnormalities incell shape or microtubule organization in SPDL-1-de-pleted embryos that resembled the defects seen follow-ing RNAi of DmSpindly in interphase S2 cells. Prelimi-nary work in human tissue culture cells indicates thathuman Spindly is similar to C. elegans SPDL-1 in that itis required to recruit both dynein and dynactin to kineto-chores, and its inhibition does not result in detectabledefects in interphase microtubule organization or cellshape. However, like DmSpindly, the human homolog isnot required for Mad2 localization or spindle checkpointactivation (R. Gassmann and A. Desai, unpubl.).

Previous studies have shown that cytoplasmic dyneinas well as its accessory factors dynactin and LIS-1 areinvolved in multiple processes during the first embry-onic division of C. elegans, including pronuclear mi-gration, centrosome separation, and bipolar spindle as-sembly (Gonczy et al. 1999; Cockell et al. 2004; Schmidtet al. 2005; O’Rourke et al. 2007). All of these processesare unaffected following SPDL-1 depletion, indicat-ing that SPDL-1 does not globally control dynein func-tion.

The C. elegans RZZ complex

The close relationship between SPDL-1 and the RZZ ho-mologs led us to functionally characterize the RZZ com-plex in C. elegans. The RZZ complex has been studiedprimarily in D. melanogaster with recent contributionsfrom vertebrate systems (Karess 2005). Our results in C.elegans confirm that ROD-1, ZwilchZWL-1, andZw10CZW-1 function as a complex that localizes tran-siently to kinetochores from NEBD until the onset ofanaphase. The RZZ complex localizes to the outer ki-netochore downstream from KNL-1 and, like SPDL-1, isnot required for kinetochore targeting of the NDC-80complex. In vertebrates, the coiled-coil protein Zwintacts as an intermediary between KNL-1 and the RZZcomplex, but Zwint-like molecules have not been iden-tified in C. elegans or D. melanogaster (Starr et al. 2000;Cheeseman et al. 2004; Kops et al. 2005). The two knownroles of the RZZ complex, recruitment of dynein/dynac-tin to unattached kinetochores and activation of thespindle checkpoint through kinetochore targeting ofMad2, are conserved in C. elegans.

Gassmann et al.

2394 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 11: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

RNAi in C. elegans suggests that Zw10CZW-1, but notROD-1 or ZwilchZWL-1, has an additional nonkineto-chore function in membrane trafficking, as previouslyreported in vertebrates (Hirose et al. 2004). This differ-ence between the RZZ subunits raises a cautionarynote about interpreting Zw10 perturbations strictlyin terms of kinetochore function, and we were carefulto focus on ROD-1 and ZwilchZWL-1 as targets to spe-cifically inhibit RZZ complex activity at kineto-chores.

Implications of C. elegans RZZ complex analysisfor the role of dynein/dynactin at the kinetochore

Chromosome movement Dynein/dynactin and the mi-crotubule-binding NDC-80 complex are independentlytargeted to kinetochores downstream from KNL-1.While the NDC-80 complex is required to make load-bearing kinetochore–microtubule attachments, its inhi-bition does not result in a “kinetochore-null” phenotypeas clear residual chromosome movements are observed

Figure 7. Codepletion of SPDL-1 orROD-1 with NDC-80 recapitulates the“kinetochore-null” phenotype. (A)Frames from time-lapse sequences repre-senting metaphase (200 sec after NEBD)and telophase (320 sec after NEBD). Co-depletion of SPDL-1 or ROD-1 withNDC-80 approximates the “kinetochore-null” phenotype of knl-3(RNAi) embryos(see also Supplemental Movies 13 and 14),in which chromosomes of the two pronu-clei are often visible as separate clumps atmetaphase (arrows), and unsegregatedchromatin remains at the spindle equatorin telophase (arrowheads). Bar, 5 µm. (B)Percentage of first divisions displayingthe chromosome morphologies describedin A at 200 sec and 320 sec after NEBD.(C) Schematic summary of the relation-ship between the RZZ complex, SPDL-1,dynein/dynactin, and the NDC-80 com-plex. The negative regulation of the KMNnetwork by the RZZ complex, which istransient in the wild-type situation, maybe either direct or indirect. (D) Model ex-plaining the difference in phenotypic se-verity between SPDL-1 and RZZ complexinhibitions. Specifically, we propose thatSPDL-1 depletion results in persistentRZZ complex-mediated inhibition of theKMN network (until just prior to ana-phase onset), because RZZ complex local-ization to kinetochores is uncoupled fromdynein/dynactin. In RZZ subunit deple-tions or codepletions of SPDL-1 withRZZ subunits, the inhibitory mechanismis absent, resulting in the weaker pheno-type, which reflects loss of dynein contri-bution to the establishment and orienta-tion of load-bearing attachments. (E) Aspeculative model for the physiologicalrole of a RZZ complex-mediated inhibi-tion of the KMN network during pro-metaphase. Dynein/dynactin laterallycaptures microtubules to accelerate for-mation of end-coupled attachments of

correct geometry. While a microtubule is laterally bound, dynein motility does not experience significant resistance (green arrow);consequently, there is low intrakinetochore tension, and the RZZ complex inhibits the KMN network from binding prematurely tothe microtubule, which would interfere with dynein-mediated kinetochore orientation. When the plus end of the microtubulebecomes embedded into the outer plate (end-coupled attachment) and provides resistance to dynein/dynactin motility (red arrow), theincreased intrakinetochore tension turns off the inhibitory action of the RZZ complex, allowing formation of stable load-bearingattachments.

Dynein function at kinetochores

GENES & DEVELOPMENT 2395

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 12: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

(Desai et al. 2003). Such Ndc80 complex-independentmovements have also been described in vertebrate cellsand dynein/dynactin has been implicated as their source(McCleland et al. 2004; DeLuca et al. 2005; Vorozhko etal. 2008). Our finding that double depletions of NDC-80with either ROD-1 or SPDL-1 synergistically recapitu-late the “kinetochore-null” phenotype is consistent withthe conclusion that both of these microtubule-bindingactivities contribute to chromosome–spindle microtu-bule interactions downstream from KNL-1.

Kinetics of load-bearing attachment formation In theabsence of the NDC-80 complex, kinetochore-localizeddynein/dynactin is insufficient to generate load-bearingattachments that can oppose the effects of aster-basedcortical pulling forces during chromosome alignment. InRZZ complex-inhibited embryos, formation of load-bearing attachments occurs, but is delayed. This resultsuggests that kinetochore-localized dynein/dynactin ac-celerates the formation of NDC-80 complex dependentend-coupled attachments. We speculate that this kineticacceleration is due to the ability of dynein/dynactin toefficiently collect microtubules that pass by the kineto-chore. Such microtubules would remain laterally associ-ated with the kinetochore until their dynamic plus endsare close enough to be integrated into the outer plate bythe KMN network. Importantly, in RZZ complex-inhib-ited embryos, despite the kinetic defect in forming load-bearing attachments, spindles always reached wild-typelength at anaphase onset and had a tightly aligned meta-phase plate. Thus, kinetochore dynein/dynactin is dis-pensable for end-coupled load-bearing attachments, butit accelerates their formation in early prometaphase.

Attachment geometry In addition to the kinetic effecton the formation of load-bearing attachments, RZZ com-plex inhibitions revealed an increased frequency of lag-ging chromatin during anaphase. Importantly, depletionof either Mad1MDF-1 or Mad2MDF-2, both essential com-ponents of the spindle checkpoint, does not have anydeleterious effects on chromosome segregation in thefirst embryonic division (A. Essex and A. Desai, in prep.).Thus, the lack of a spindle checkpoint-mediated cellcycle delay cannot explain the chromosome missegrega-tion observed in RZZ complex inhibitions. This findingis similar to the situation in D. melanogaster, wheremad2-null mutants are reported to have little or no chro-mosome segregation defects, while the signature pheno-type of zw10- and rod-null mutants is lagging anaphasechromatin (Karess and Glover 1989; Williams et al. 1992;Buffin et al. 2007). Since RZZ complex-inhibited em-bryos have no noticeable defects in chromosome conden-sation, it is likely that the chromatin bridges in anaphaseare caused by incorrect merotelic attachments, where asingle kinetochore is connected to both poles (Cimini etal. 2003). We propose that a major role of kinetochoredynein/dynactin is to prevent the generation of suchmaloriented kinetochores in early prometaphase, whenkinetochore–microtubule interactions are first estab-lished. When an unattached sister kinetochore binds lat-

erally to an astral microtubule, the minus-end-directedmotility of dynein/dynactin would provide a force thatorients the kinetochore toward the spindle pole at whichthat particular microtubule originates, thereby decreas-ing the probability that the same kinetochore captures amicrotubule from the opposite pole. Thus, dynein/dyn-actin would ensure correct attachment geometry by forc-ing sister kinetochores to face opposite poles.

Transient inhibition of load-bearing kinetochore–microtubule attachments by the RZZ complex:a mechanism to coordinate the transitionfrom lateral to end-coupled attachments?

The RZZ complex and SPDL-1 are equivalently requiredfor both dynein/dynactin targeting to kinetochores andspindle checkpoint activation. Yet, the consequences oftheir inhibition are strikingly different. Pole trackinganalysis revealed that the consequences of inhibitingSPDL-1 are indistinguishable from complete loss ofload-bearing attachments until just prior to anaphase on-set. In contrast, in the RZZ complex inhibitions, after aslight delay, kinetochore–microtubule attachmentseventually bear load equally well as those of unperturbedembryos, resulting in wild-type spindle length at ana-phase onset.

This comparison reveals that following SPDL-1 inhi-bition there is a significant defect in formation of KMNnetwork-mediated load-bearing attachments. The defectis attributable to the presence of the RZZ complex atkinetochores, as coinhibition of SPDL-1 and the RZZcomplex reduces the phenotypic severity to match thatof inhibiting the RZZ complex alone. This observationindicates that the RZZ complex negatively regulatesKMN network activity and that this negative regulationpersists for most of prometaphase in the absence ofSPDL-1 (Fig. 7C,D). The RZZ complex may inhibit theKMN network either directly or via other regulators ofKMN network function. Aurora B kinase is known tonegatively regulate the microtubule-binding activity ofthe Ndc80 complex (Cheeseman et al. 2006; DeLuca etal. 2006). In preliminary work using a temperature-sen-sitive Aurora BAIR-2 mutant, coinhibition of Aurora B didnot reduce the severity of the chromosome segregationdefect following SPDL-1 depletion (R. Gassmann and A.Desai, unpubl.), suggesting that the RZZ complex doesnot regulate the KMN network through Aurora B. In theabsence of the RZZ complex from kinetochores, themechanism for negatively regulating KMN network ac-tivity is no longer present, explaining why coinhibitionof SPDL-1 and the RZZ complex quantitatively reducesthe phenotypic severity to match that of the RZZ com-plex alone (Fig. 7D).

We propose that the physiological function of the regu-latory link between the RZZ complex and the KMN net-work is to ensure a coordinated transition from transientlateral attachments made by dynein, which accelerateformation of end-coupled attachments of correct geom-etry, to stable load-bearing end-coupled attachmentsthat do the job of chromosome segregation. In this

Gassmann et al.

2396 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 13: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

model, RZZ complex inhibition of the KMN network ismodulated by the microtubule minus-end-directed mo-tility of dynein/dynactin, which is linked to the outerkinetochore via SPDL-1 and the RZZ complex (Fig. 7E).When dynein/dynactin is laterally attached to a micro-tubule that extends past the kinetochore, the RZZ com-plex is under low tension and negatively regulates themicrotubule-binding activity of the KMN network. Thisprevents the KMN network from tightly binding to amicrotubule extending past the kinetochore that hasbeen captured by dynein/dynactin. When dynein/dynac-tin translocation toward the microtubule minus end ismet with resistance due to the microtubule plus endbeing embedded in the kinetochore outer plate, the RZZcomplex is placed under tension and inhibition of theKMN network is relieved. We envision that such a feed-back mechanism prevents premature lateral binding ofthe KMN network to microtubules, which would inter-fere with dynein-mediated kinetochore orientation andincrease the likelihood of forming incorrect merotelicattachments.

In summary, our results reveal a new mechanism regu-lating kinetochore–microtubule attachments that in-volves the RZZ complex and is likely to be modulated bydynein/dynactin activity. We speculate on the underly-ing reason for why such a regulatory mechanism wouldbe necessary for the fidelity of chromosome segregation.This mechanism is likely to be integrated with spindlecheckpoint signaling that also requires the RZZ complexin all metazoans.

Materials and methods

Worm strains and antibodies

Worm strains used in this study are listed in SupplementalTable 1. For worm GFPLAP fusions of SPDL-1, ZWL-1, CZW-1,and MDF-2, the genomic locus was cloned into pIC26 (Cheese-man et al. 2004); the genomic locus of DNC-2 was cloned intopAZ132 (Praitis et al. 2001). For DHC-1, the start codon wasreplaced with GFP by recombineering a full-length dhc-1 fosmidclone (details will be described elsewhere). All constructs wereintegrated into the DP38 strain [unc-119(ed3)] using micropar-ticle bombardment (Praitis et al. 2001) with a PDS-1000/HeBiolistic Particle Delivery System (Bio-Rad), and mCherry:histone H2B was subsequently introduced by mating (Green etal. 2008). Affinity-purified polyclonal antibodies against full-length SPDL-1 and ZwilchZWL-1 were generated as describedpreviously (Desai et al. 2003).

RNAi

L4 worms were injected with dsRNA (Supplemental Table 2)prepared as described previously (Oegema et al. 2001) and incu-bated for 48 h at 20°C. For double depletions, dsRNAs weremixed to obtain equal concentrations of �0.75 mg/mL for eachdsRNA.

Immunofluorescence

For stainings with the anti-SPDL-1 antibody, embryos werefixed for 5 min in 3% paraformaldehyde as detailed previously

(Howe et al. 2001). Immunofluorescence for other antibodiesand microscopy was performed as described in Oegema et al.(2001) and Cheeseman et al. (2004), respectively. All antibodiesused were directly labeled with fluorescent dyes (Cy2, Cy3, orCy5; Amersham Biosciences).

Live imaging

Time-lapse movies of worm strain TH32 (coexpressing GFP:histone H2B and GFP:�-tubulin) (Oegema et al. 2001) were ac-quired at 21°C on a Nikon Eclipse E800 microscope using acharge-coupled device camera (Orca-ER; Hamamatsu Photon-ics) at 2 × 2 binning, and a 60× 1.4 NA Plan Apochromat objec-tive. Acquisition parameters, shutters, and focus were con-trolled by MetaMorph software (MDS Analytical Technologies).Quantitative analysis of spindle pole elongation was performedusing a MetaMorph algorithm (Desai et al. 2003). Movies ofstrain TH32 for the spindle checkpoint assay were recorded at18°C on a DeltaVision microscope (Applied Precision) equippedwith a CoolSnap charge-coupled device camera (Roper Scien-tific) at 2 × 2 binning and a 100× NA 1.3 U-planApo objective(Olympus). Imaging of all other worm strains was performed at21°C on a spinning disc confocal head (McBain Instruments)mounted on an inverted Nikon TE2000e microscope equippedwith a 60× 1.4 NA Plan Apochromat lens (Nikon), a krypton-argon 2.5 W water-cooled laser (Spectra-Physics), and a charge-coupled device camera (iXon; Andor Technology, or Orca-ER;Hamamatsu Photonics). Acquisition parameters, shutters, andfocus were controlled by MetaMorph software. Imaging condi-tions for individual strains are listed in Supplemental Table 3.

Immunoprecipitations, LAP purifications, and massspectrometry

Immunoprecipitations were conducted on high-speed superna-tant from adult worms as described previously (Cheeseman etal. 2004). For Western blotting, proteins were eluted from theantibody–Protein A resin with sample buffer (50 mM Tris-HClat pH 6.8, 15% [w/v] sucrose, 2 mM EDTA, 3% SDS) for 15 minat 70°C. For mass spectrometric analysis, the elution was per-formed with 8 M urea in 50 mM Tris-HCl (pH 8.5). LAP puri-fication of ZwilchZWL-1 and mass spectrometry were conductedas described previously (Cheeseman et al. 2001, 2004). Tandemmass spectra were searched against the most recent version ofthe predicted C. elegans proteins (Wormpep111).

Acknowledgments

We are grateful to Ana Carvalho, Andrew Holland, and AlexDammermann for critical reading of the manuscript. This workwas supported by a National Science Foundation of Switzerlandfellowship (to R.G.), the UCSD Genetics Training Grant (toA.E.), a Leukemia and Lymphoma Society fellowship (to S.O.), aDamon Runyon Cancer Research Foundation fellowship (toP.M.), grants from the NIH to A.D. (GM074215) and to B.B(GM49869), a Scholar Award from the Damon Runyon CancerResearch Foundation to A.D. (DRS 38-04), and funding from theLudwig Institute for Cancer Research to A.D. and K.O.

References

Basto, R., Gomes, R., and Karess, R.E. 2000. Rough deal andZw10 are required for the metaphase checkpoint in Dro-sophila. Nat. Cell Biol. 2: 939–943.

Buffin, E., Lefebvre, C., Huang, J., Gagou, M.E., and Karess, R.E.

Dynein function at kinetochores

GENES & DEVELOPMENT 2397

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 14: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

2005. Recruitment of Mad2 to the kinetochore requires theRod/Zw10 complex. Curr. Biol. 15: 856–861.

Buffin, E., Emre, D., and Karess, R.E. 2007. Flies without aspindle checkpoint. Nat. Cell Biol. 9: 565–572.

Chan, G.K., Jablonski, S.A., Starr, D.A., Goldberg, M.L., andYen, T.J. 2000. Human Zw10 and ROD are mitotic check-point proteins that bind to kinetochores. Nat. Cell Biol. 2:944–947.

Cheeseman, I.M., Brew, C., Wolyniak, M., Desai, A., Anderson,S., Muster, N., Yates, J.R., Huffaker, T.C., Drubin, D.G., andBarnes, G. 2001. Implication of a novel multiprotein Dam1pcomplex in outer kinetochore function. J. Cell Biol. 155:1137–1145.

Cheeseman, I.M., Niessen, S., Anderson, S., Hyndman, F., YatesIII, J.R., Oegema, K., and Desai, A. 2004. A conserved proteinnetwork controls assembly of the outer kinetochore and itsability to sustain tension. Genes & Dev. 18: 2255–2268.

Cheeseman, I.M., MacLeod, I., Yates III, J.R., Oegema, K., andDesai, A. 2005. The CENP-F-like proteins HCP-1 and HCP-2target CLASP to kinetochores to mediate chromosome seg-regation. Curr. Biol. 15: 771–777.

Cheeseman, I.M., Chappie, J.S., Wilson-Kubalek, E.M., and De-sai, A. 2006. The conserved KMN network constitutes thecore microtubule-binding site of the kinetochore. Cell 127:983–997.

Cimini, D., Moree, B., Canman, J.C., and Salmon, E.D. 2003.Merotelic kinetochore orientation occurs frequently duringearly mitosis in mammalian tissue cells and error correctionis achieved by two different mechanisms. J. Cell Sci. 116:4213–4225.

Cockell, M.M., Baumer, K., and Gonczy, P. 2004. lis-1 is re-quired for dynein-dependent cell division processes in C. el-egans embryos. J. Cell Sci. 117: 4571–4582.

DeLuca, J.G., Dong, Y., Hergert, P., Strauss, J., Hickey, J.M.,Salmon, E.D., and McEwen, B.F. 2005. Hec1 and nuf2 arecore components of the kinetochore outer plate essential fororganizing microtubule attachment sites. Mol. Biol. Cell 16:519–531.

DeLuca, J.G., Gall, W.E., Ciferri, C., Cimini, D., Musacchio, A.,and Salmon, E.D. 2006. Kinetochore microtubule dynamicsand attachment stability are regulated by Hec1. Cell 127:969–982.

Desai, A., Rybina, S., Muller-Reichert, T., Shevchenko, A.,Shevchenko, A., Hyman, A., and Oegema, K. 2003. KNL-1directs assembly of the microtubule-binding interface of thekinetochore in C. elegans. Genes & Dev. 17: 2421–2435.

Gonczy, P., Pichler, S., Kirkham, M., and Hyman, A.A. 1999.Cytoplasmic dynein is required for distinct aspects ofMTOC positioning, including centrosome separation, in theone cell stage Caenorhabditis elegans embryo. J. Cell Biol.147: 135–150.

Green, R.A., Audhya, A., Pozniakovsky, A., Dammermann, A.,Pemble, H., Monen, J., Portier, N., Hyman, A., Desai, A., andOegema, K. 2008. Expression and imaging of fluorescent pro-teins in the C. elegans gonad and early embryo. MethodsCell Biol. 85: 179–218.

Griffis, E.R., Stuurman, N., and Vale, R.D. 2007. Spindly, anovel protein essential for silencing the spindle assemblycheckpoint, recruits dynein to the kinetochore. J. Cell Biol.177: 1005–1015.

Hirose, H., Arasaki, K., Dohmae, N., Takio, K., Hatsuzawa, K.,Nagahama, M., Tani, K., Yamamoto, A., Tohyama, M., andTagaya, M. 2004. Implication of ZW10 in membrane traf-ficking between the endoplasmic reticulum and Golgi.EMBO J. 23: 1267–1278.

Howe, M., McDonald, K.L., Albertson, D.G., and Meyer, B.J.

2001. HIM-10 is required for kinetochore structure and func-tion on Caenorhabditis elegans holocentric chromosomes. J.Cell Biol. 153: 1227–1238.

Howell, B.J., McEwen, B.F., Canman, J.C., Hoffman, D.B., Far-rar, E.M., Rieder, C.L., and Salmon, E.D. 2001. Cytoplasmicdynein/dynactin drives kinetochore protein transport to thespindle poles and has a role in mitotic spindle checkpointinactivation. J. Cell Biol. 155: 1159–1172.

Karess, R. 2005. Rod–Zw10–Zwilch: A key player in the spindlecheckpoint. Trends Cell Biol. 15: 386–392.

Karess, R.E. and Glover, D.M. 1989. rough deal: A gene requiredfor proper mitotic segregation in Drosophila. J. Cell Biol.109: 2951–2961.

Kline-Smith, S.L., Sandall, S., and Desai, A. 2005. Kinetochore–spindle microtubule interactions during mitosis. Curr.Opin. Cell Biol. 17: 35–46.

Kops, G.J., Kim, Y., Weaver, B.A., Mao, Y., McLeod, I., Yates III,J.R., Tagaya, M., and Cleveland, D.W. 2005. ZW10 links mi-totic checkpoint signaling to the structural kinetochore. J.Cell Biol. 169: 49–60.

Li, Y., Yu, W., Liang, Y., and Zhu, X. 2007. Kinetochore dyneingenerates a poleward pulling force to facilitate congressionand full chromosome alignment. Cell Res. 17: 701–712.

Maddox, P.S., Hyndman, F., Monen, J., Oegema, K., and Desai,A. 2007. Functional genomics identifies a Myb domain-con-taining protein family required for assembly of CENP-Achromatin. J. Cell Biol. 176: 757–763.

McCleland, M.L., Kallio, M.J., Barrett-Wilt, G.A., Kestner, C.A.,Shabanowitz, J., Hunt, D.F., Gorbsky, G.J., and Stukenberg,P.T. 2004. The vertebrate Ndc80 complex contains Spc24and Spc25 homologs, which are required to establish andmaintain kinetochore–microtubule attachment. Curr. Biol.14: 131–137.

Musacchio, A. and Salmon, E.D. 2007. The spindle-assemblycheckpoint in space and time. Nat. Rev. Mol. Cell Biol. 8:379–393.

Nicklas, R.B. 1988. The forces that move chromosomes in mi-tosis. Annu. Rev. Biophys. Biophys. Chem. 17: 431–449.

O’Connell, K.F., Caron, C., Kopish, K.R., Hurd, D.D., Kem-phues, K.J., Li, Y., and White, J.G. 2001. The C. elegans zyg-1gene encodes a regulator of centrosome duplication with dis-tinct maternal and paternal roles in the embryo. Cell 105:547–558.

Oegema, K., Desai, A., Rybina, S., Kirkham, M., and Hyman,A.A. 2001. Functional analysis of kinetochore assembly inCaenorhabditis elegans. J. Cell Biol. 153: 1209–1226.

O’Rourke, S.M., Dorfman, M.D., Carter, J.C., and Bowerman, B.2007. Dynein modifiers in C. elegans: Light chains suppressconditional heavy chain mutants. PLoS Genet. 3: e128. doi:10.1371/journal.pgen.0030128.

Praitis, V., Casey, E., Collar, D., and Austin, J. 2001. Creation oflow-copy integrated transgenic lines in Caenorhabditis el-egans. Genetics 157: 1217–1226.

Rieder, C.L. and Alexander, S.P. 1990. Kinetochores are trans-ported poleward along a single astral microtubule duringchromosome attachment to the spindle in newt lung cells. J.Cell Biol. 110: 81–95.

Savoian, M.S., Goldberg, M.L., and Rieder, C.L. 2000. The rateof poleward chromosome motion is attenuated in Dro-sophila zw10 and rod mutants. Nat. Cell Biol. 2: 948–952.

Scaerou, F., Aguilera, I., Saunders, R., Kane, N., Blottiere, L., andKaress, R. 1999. The rough deal protein is a new kinetochorecomponent required for accurate chromosome segregation inDrosophila. J. Cell Sci. 112: 3757–3768.

Scaerou, F., Starr, D.A., Piano, F., Papoulas, O., Karess, R.E., andGoldberg, M.L. 2001. The ZW10 and Rough Deal checkpoint

Gassmann et al.

2398 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 15: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

proteins function together in a large, evolutionarily con-served complex targeted to the kinetochore. J. Cell Sci. 114:3103–3114.

Schmidt, D.J., Rose, D.J., Saxton, W.M., and Strome, S. 2005.Functional analysis of cytoplasmic dynein heavy chain inCaenorhabditis elegans with fast-acting temperature-sensi-tive mutations. Mol. Biol. Cell 16: 1200–1212.

Smith, D.A., Baker, B.S., and Gatti, M. 1985. Mutations in genesencoding essential mitotic functions in Drosophila melano-gaster. Genetics 110: 647–670.

Sonnichsen, B., Koski, L.B., Walsh, A., Marschall, P., Neumann,B., Brehm, M., Alleaume, A.M., Artelt, J., Bettencourt, P.,Cassin, E., et al. 2005. Full-genome RNAi profiling of earlyembryogenesis in Caenorhabditis elegans. Nature 434: 462–469.

Starr, D.A., Williams, B.C., Hays, T.S., and Goldberg, M.L. 1998.ZW10 helps recruit dynactin and dynein to the kinetochore.J. Cell Biol. 142: 763–774.

Starr, D.A., Saffery, R., Li, Z., Simpson, A.E., Choo, K.H., Yen,T.J., and Goldberg, M.L. 2000. HZwint-1, a novel humankinetochore component that interacts with HZW10. J. CellSci. 113: 1939–1950.

Stehman, S.A., Chen, Y., McKenney, R.J., and Vallee, R.B. 2007.NudE and NudEL are required for mitotic progression andare involved in dynein recruitment to kinetochores. J. CellBiol. 178: 583–594.

Vergnolle, M.A. and Taylor, S.S. 2007. Cenp-F links kineto-chores to Ndel1/Nde1/Lis1/dynein microtubule motor com-plexes. Curr. Biol. 17: 1173–1179.

Vorozhko, V.V., Emanuele, M.J., Kallio, M.J., Stukenberg, P.T.,and Gorbsky, G.J. 2008. Multiple mechanisms of chromo-some movement in vertebrate cells mediated through theNdc80 complex and dynein/dynactin. Chromosoma 117:169–179.

Wei, R.R., Al-Bassam, J., and Harrison, S.C. 2007. The Ndc80/HEC1 complex is a contact point for kinetochore–microtu-bule attachment. Nat. Struct. Mol. Biol. 14: 54–59.

Williams, B.C. and Goldberg, M.L. 1994. Determinants of Dro-sophila zw10 protein localization and function. J. Cell Sci.107: 785–798.

Williams, B.C., Karr, T.L., Montgomery, J.M., and Goldberg,M.L. 1992. The Drosophila l(1)zw10 gene product, requiredfor accurate mitotic chromosome segregation, is redistribut-ed at anaphase onset. J. Cell Biol. 118: 759–773.

Williams, B.C., Li, Z., Liu, S., Williams, E.V., Leung, G., Yen,T.J., and Goldberg, M.L. 2003. Zwilch, a new component ofthe ZW10/ROD complex required for kinetochore functions.Mol. Biol. Cell 14: 1379–1391.

Wojcik, E., Basto, R., Serr, M., Scaerou, F., Karess, R., and Hays,T. 2001. Kinetochore dynein: Its dynamics and role in thetransport of the Rough deal checkpoint protein. Nat. CellBiol. 3: 1001–1007.

Yang, Z., Tulu, U.S., Wadsworth, P., and Rieder, C.L. 2007.Kinetochore dynein is required for chromosome motion andcongression independent of the spindle checkpoint. Curr.Biol. 17: 973–980.

Dynein function at kinetochores

GENES & DEVELOPMENT 2399

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from

Page 16: A new mechanism controlling kinetochore–microtubule ...genesdev.cshlp.org/content/22/17/2385.full.pdf · Asako Sugimoto,2 Sean M. O’Rourke,3 Bruce Bowerman,3 Ian McLeod,4 John

10.1101/gad.1687508Access the most recent version at doi: 22:2008, Genes Dev. 

  Reto Gassmann, Anthony Essex, Jia-Sheng Hu, et al.   and the Rod/Zwilch/Zw10 complexrevealed by comparison of two dynein-targeting components: SPDL-1

microtubule interactions−A new mechanism controlling kinetochore

  Material

Supplemental 

http://genesdev.cshlp.org/content/suppl/2008/08/14/22.17.2385.DC1

Related Content

  Genes Dev. September , 2008 22: 2302-2307

Filiz Çivril and Andrea MusacchioSpindly attachments

  References

  http://genesdev.cshlp.org/content/22/17/2385.full.html#related-urls

Articles cited in: 

http://genesdev.cshlp.org/content/22/17/2385.full.html#ref-list-1This article cites 50 articles, 27 of which can be accessed free at:

  License

ServiceEmail Alerting

  click here.right corner of the article or

Receive free email alerts when new articles cite this article - sign up in the box at the top

Copyright © 2008, Cold Spring Harbor Laboratory Press

Cold Spring Harbor Laboratory Press on January 13, 2021 - Published by genesdev.cshlp.orgDownloaded from


Recommended