<<

Current , Vol. 15, R265–R275, April 12, 2005, ©2005 Elsevier Ltd. All rights reserved. DOI 10.1016/j.cub.2005.03.037

Condensins: Organizing and Review Segregating the

Tatsuya Hirano and mechanisms behind this scene have largely remained elusive. During the past decade, however, a substantial body of evidence has accumulated that a are multi-subunit complexes that class of multiprotein complexes known as condensins play a central role in mitotic assembly is one of the key components that directly regulates and segregation. The complexes contain ‘structural chromosome architecture and dynamics. The chief maintenance of ’ (SMC) ATPase sub- function of condensins is to assemble and segregate units, and induce DNA supercoiling and looping in an chromosomes during and meiosis, though ATP--dependent manner in vitro. recent studies suggest that they also play much cells have two different complexes, con- broader roles in the expression and maintenance of densins I and II, each containing a unique set of reg- the genome. In this review, I will summarize recent ulatory subunits. Condensin II participates in an early progress in our understanding of the structure and stage of chromosome condensation within the function of condensins, and discuss unresolved prob- nucleus. Condensin I gains access to chro- lems and future directions in the field. mosomes only after the breaks down, and collaborates with condensin II to assemble Subunit Composition and Molecular Architecture of chromosomes with fully resolved sister Condensins chromatids. The complexes also play critical roles in The canonical condensin complex, now referred to as meiotic and in condensin I, was originally identified as a major processes such as repression and checkpoint protein component required for the establishment and responses. In bacterial cells, ancestral forms of con- maintenance of mitotic chromosomes in -free densins control chromosome dynamics. Dissecting extracts of Xenopus laevis [1,2]. More recently, a the diverse functions of condensins is likely to be second condensin complex, condensin II, has been central to our understanding of genome organization, found in vertebrate cells [3,4]. Condensins I and II stability and . have the same pair of core subunits but distinct sets of regulatory subunits. The two core subunits, CAP- E/SMC2 and CAP-C/SMC4, belong to a large family of chromosomal ATPases known as the ‘structural main- Introduction tenance of chromosomes’ (SMC) family. The In this era of genomics, a huge amount of sequence SMC2–SMC4 heterodimer adopts a V-shaped struc- information is accumulating every day, and the ture, characteristic of all SMC , with an ATP- genome content of many species is being uncovered binding cassette (ABC) ATPase at the distal at an extraordinary pace. One of the important chal- end of each arm (Figure 1A,i) [5,6]. Like other SMC lenges in post-genomics research is to understand proteins, it is predicted that two ATP are how the genome is organized into three-dimensional sandwiched between the ATP-binding ‘head’ domains structure within the cell. It is also important [7,8], and that ATP binding and hydrolysis modulate to determine how such structure is remodeled and engagement and disengagement of the head modified during the duplication, expression and domains, respectively (Figure 1A,ii) [9,10]. How the inheritance of the genome. Cytologically, the most mechanochemical cycle of the SMC subunits is used striking transformation of chromatin structure is to reconfigure DNA and chromatin structure is under observed during mitosis when the cell partitions its active investigation (see below). Note that the SMC duplicated genome into daughter cells. Upon disas- heterodimer is a huge : each arm, composed sembly of the cell nucleus at the onset of mitosis, an of an anti-parallel , is about 50 nm long, a amorphous mass of chromatin is converted into a length equivalent to that of 150 base pairs of double- discrete set of rod-shaped chromosomes in which stranded DNA. the two duplicated ‘sister’ chromatids are juxtaposed The two different condensin complexes are distin- along their length. This process, referred to as chro- guished by their unique sets of non-SMC subunits. mosome assembly or condensation, is an essential Among the three regulatory subunits of each complex, prerequisite of the faithful segregation of sister chro- two — CAP-D2 and CAP-G in condensin I, and CAP- matids at the subsequent stage of mitosis D3 and CAP-G2 in condensin II — contain HEAT (anaphase). repeats, a highly degenerate repeat motif implicated in Although the sudden appearance of thread-like protein–protein interactions [11]. The fifth subunits — chromosomes in early mitosis has fascinated cell biol- CAP-H in condensin I and CAP-H2 in condensin II — ogists for more than 100 years, the molecular players belong to the kleisin family of proteins [12]. The non- SMC subunits of condensin I form a subcomplex that Cold Spring Harbor Laboratory, One Bungtown Road, P.O. binds to the head domain(s) of the SMC heterodimer Box 100, Cold Spring Harbor, New York 11724, USA. (Figure 1B,i) [6,13]. It has been shown that the non- E-mail: [email protected] SMC subunits regulate chromosomal targeting of the Review R266

Figure 1. Architecture of condensins A (i) (ii) and their related complexes. Hinge (A) SMC2 and SMC4 form the core of ATP the condensin complexes. Each SMC binding subunit folds intramolecularly by anti- N C N C Coiled-coil C N C N parallel coiled-coil interactions, and arm forms an ATP-binding head domain composed of its amino- and carboxy- ATP Engagement hydrolysis terminal sequences. A hinge–hinge interaction between SMC2 and SMC4 N NN C then produces a V-shaped heterodimer CCHead C N (i). Two ATP molecules (yellow) are SMC2 SMC4 sandwiched between two SMC head domains and induce their engagement. Disengagement of the two head B (i)(ii) (iii) (iv) domains is triggered upon hydrolysis of the ATP (ii). (B) Condensin I (i) and con- densin II (ii) share the same pair of SMC2 and SMC4 as their core sub- units. Each of the three non-SMC sub- SMC2 SMC4 SMC2 SMC4 SMC2 SMC4V SMC SMC units of condensin I has a distantly (DPY-27) related counterpart in those of con- densin II. The CAP-D2, CAP-G, CAP-D3 and CAP-G2 subunits contain HEAT H H2 DPY-26 repeats, whereas the CAP-H and CAP- ScpA H2 subunits belong to the kleisin family of proteins. The nematode Caenorhab- D2 G D3 G2 DPY-28 ScpB ditis elegans has a specialized con- densin-like complex which regulates Condensin I Condensin II Dosage Bacterial dosage compensation (iii). The core of compensation SMC complex this complex is a heterodimer of the complex authentic SMC2 subunit and an SMC4 variant called DPY-27. DPY-28 has kleisin HEAT HEAT repeats whereas DPY-26 has Current Biology kleisin motifs. The Bacillus subtilis SMC is composed of an SMC homodimer, a kleisin subunit (ScpA) and another small subunit called ScpB (iv).

SMC dimer in Xenopus extracts and modulate its organization (Figure 2A and B), the of an ATPase and DNA-binding activities in vitro [14]. Con- is not always a good indicator to predict densin II is anticipated to have a similar architecture whether it possesses condensin II or not. For (Figure 1B,ii). example, all condensin II were found in the Eukaryotic cells have another SMC protein genome of the unicellular red Cyanidioschyzon complex, known as , that plays a central role merolae (Table 1) [16]: this compact genome is only in sister chromatid cohesion during mitosis and 16.5 Mb, a similar size to that of the meiosis [15]. The core of the cohesin complex is a Saccharomyces cerevisiae. heterodimer of SMC1 and SMC3, and one of the two The apparent loss of condensin I in C. elegans and regulatory subunits (Scc1) belongs to the kleisin C. briggsae is puzzling, but may be related to their family. Despite their similar subunit composition, the unique, holocentric chromosome structure (Figure cohesin and condensin complexes display different 2C) [17,18]. Alternatively, an ancient condensin I arm conformations, as judged by electron complex may have lost its mitotic functions during microscopy, which may contribute to their specialized evolution of these nematodes and been modified and cellular and biochemical functions [6]. adapted to execute a specialized function in the dosage compensation that equalizes expression of X- Phylogeny of Condensin Subunits linked genes in the two sexes [19]. The core of the Phylogenic analysis of condensin subunits sheds dosage compensation complex (DCC) is composed intriguing light on the evolution of chromosome archi- of the SMC2 ortholog MIX-1 and an SMC4 variant tecture. All non-SMC subunits of condensin I are (DPY-27) that is unique to the worm. The complex highly conserved from yeast to humans, with notable also contains two non-SMC subunits, DPY-28 and exceptions of the nematodes Caenorhabditis elegans DPY-26, which have limited similarities to the CAP-D2 and C. briggsae (Table 1). The non-SMC subunits of and CAP-H subunits of condensin I, respectively condensin II are found in and , but (Figure 1B,iii; Table 1). not in yeast or other fungi such as Aspergillus nidu- Finally, it is important to note that even bacterial cells lans and Neurospora crassa. While it is tempting to have condensin-like complexes. In Bacillus subtilis, for speculate that condensin II has evolved to provide example, an SMC homodimer associates with two non- large chromosomes with an additional level of SMC subunits, ScpA and ScpB, and participates in Current Biology R267

Table 1. Condensin subunits in model .

Subunits Human Xenopus Arabidopsis Drosophila C. elegans C. merolae S. pombe S. cerevisiae (3,080 Mb)1 (125 Mb) (165 Mb) (100 Mb) (16.5 Mb) (13.8 Mb) (12.1 Mb)

Core (I & II) DCC2 SMC hCAP-E XCAP-E AtCAP-E1/E2 DmSMC2 MIX-1 MIX-1 CMG189C Cut14 Smc2

SMC hCAP-C XCAP-C AtCAP-C DmSMC4 SMC-4 (DPY-27) CME029C Cut3 Smc4

I-specific HEAT hCAP-D2 XCAP-D2 CAB72176 CG1911 – (DPY-28) CMR484C Cnd1 Ycs4

HEAT hCAP-G XCAP-G BAB08309 CG17054 – – CMS422C Cnd3 Ycs5/Ycg1 kleisin hCAP-H XCAP-H AAC25941 Barren – (DPY-26) CMF069C Cnd2 Brn1

II-specific HEAT hCAP-D3 XCAP-D3 At4g15890.1 CG31989 HCP-6 CM0236C – –

HEAT hCAP-G2 XCAP-G2 At1g64960.1 – F55C5.4 CMA089C – – kleisin hCAP-H2 XCAP-H2 At3g16730.1 CG14685 C29E4.2 CMI207C – –

1The genome size of each organism is shown in parentheses. 2The subunits of the C. elegans dosage compensation complex (DCC) are boxed. MIX-1 is the authentic SMC2 whereas DPY-27 is a variant of SMC4. Although DPY-28 and DPY-26 are mostly related to the CAP-D2 and CAP- H subunits, respectively, they cannot be considered their orthologs. proper organization and segregation of the genome assays. It will also be important to determine whether (Figure 1B,iv) [10,20]. a single condensin complex is capable of mediating these reactions or whether cooperative interactions Molecular Actions of Condensins of multiple condensin complexes may be essential. How does the uniquely designed, two-armed struc- Full reconstitution of condensins I and II from ture of condensins interact with DNA and manipulate their recombinant subunits will be crucial to further its conformation? Despite the exciting data that are dissect the action of these highly elaborated protein accumulating, the answer to this question remains machines. elusive. Purified SMC2–SMC4 dimers have the ability Recent studies of bacterial SMC protein complexes to convert complementary single-stranded into have shed new light on the basic mechanism of action a double-stranded DNA in an ATP-independent of SMC proteins in general. By constructing mutant manner [21]. This reannealing activity appears to be forms of B. subtilis SMC proteins that are defective in supported by dynamic protein–protein interactions its ATPase cycle at three different stages, it was as judged by atomic force electron microscopy [22]. shown that ATP-dependent engagement of the head A simple gel-shift also indicates a rather domains is indeed crucial for dynamic interactions complex, cooperative interaction between the SMC between SMC proteins and DNA [10]. The non-SMC dimer and double-stranded DNA [23]. No ATP- subunits, ScpA and ScpB, play both positive and neg- dependent activities have been detected for ative roles in the SMC–DNA interactions by suppress- SMC2–SMC4 dimers, except that they display a ing the ability of the SMC complex to hydrolyze ATP. weak ATPase activity [14,23]. In Escherichia coli, the MukBEF complex is thought The holocomplex of condensin I purified from to be the structural and functional homolog of the B. Xenopus egg extracts has the ability to induce subtilis SMC–ScpAB complex. The results of single- positive superhelical tension into double-stranded DNA-molecule experiments using optical tweezers DNA in an ATP-hydrolysis-dependent manner suggest that MukBEF compacts the DNA molecule by [24–26]. Importantly, the supercoiling activity is not assembling a flexible filament in an ATP- supported by the SMC2–SMC4 heterodimer alone dependent manner [29]. ATP hydrolysis is not required [14]: it requires the non-SMC subunits and their phos- for this compaction reaction, a property significantly phorylation by the master mitotic cyclin different from that of the eukaryotic condensin I B–Cdk1 [25,27]. A similar activity has been detected complex [28]. Moreover, no supercoiling activity has in a condensin fraction purified from C. elegans been detected so far in these bacterial SMC or Muk embryos, which is predicted to be composed of con- protein complexes. Thus, it is likely that the action of densin II [18]. Most recently, a single-DNA-molecule the bacterial counterparts is related, but not identical, nanomanipulation technique using magnetic tweez- to that of the eukaryotic condensin complexes; never- ers has shown that condensin I is able to physically theless, the simple composition and ease of purification compact DNA in an ATP-hydrolysis-dependent of the former will continue to make great contributions manner [28]. The compaction reaction occurs in a to unveiling the dynamic behavior of SMC proteins. highly dynamic and reversible fashion, possibly involving a looping mechanism. It remains to be clar- Cell-Cycle Regulation of Condensins ified how this activity detected in the single-DNA-mol- Spatial and temporal distribution of the condensin sub- ecule assay is mechanistically related with the units during the appears to vary among dif- supercoiling activity revealed by the bulk biochemical ferent . For example, they are constitutively Review R268

mitotic progression. Cyclin A accumulates in the AB C nucleus during and prophase, and is degraded in late prophase or early (Figure 3B); cyclin B, however, is sequestered into the during G2 and enters the nucleus only after early prophase. Active cyclin B–Cdk1 then triggers nuclear envelope breakdown (NEBD), allowing con- densin I to gain access to chromosomes. One possi- bility is that cyclin A–Cdk1 phosphorylates and Condensin I Condensin I Condensin II activates condensin II to initiate an early stage of Condensin II chromosome condensation within the prophase Animals and plants Nematodes nucleus. Upon NEBD, cyclin B–Cdk1 phosphorylates (monocentric) (monocentric) (holocentric) and activates condensin I [25,27], and may further Current Biology phosphorylate condensin II (Figure 3C). This hypothe- sis, although admittedly highly speculative at present, explains nicely how the sequential activation of the Figure 2. Condensins and evolution of chromosome architecture. two major mitotic might contribute to spatial (A) Yeasts (S. cerevisiae and S. pombe) and other fungi and temporal regulation of chromosome condensation (A. nidulans and N. crassa) have only condensin I (blue). The chromosome is shown in light green and the sister kineto- supported by the two condensation machineries [33]. chores in yellow. are shown in gray lines. (B) Vertebrates and plants have both condensin I (blue) and Dynamic Distribution of Condensins During Mitosis condensin II (magenta). The two complexes apparently alter- A recent study [38] which combined light and electron nate along the arm and display specialized geometry at the microscopy suggests that, in mammalian /kinetochore region. (C) Nematodes (C. elegans culture cells, an early stage of chromosome conden- and C. briggsae) have condensin II (magenta), but apparently not condensin I. Unlike many other organisms in which each sation in prophase is mediated by successive coiling chromatid has a single kinetochore (monocentric), numerous of chromatin fibers: that is, hierarchical folding. kinetochores assemble along the entire length of each chro- SMC2, an SMC core subunit common to condensins matid in C. elegans (holocentric). The C. elegans condensin II I and II, associates first with peripheral regions of complex localizes to the centromere/kinetochore regions of chromosomes at this stage, and then progressively the metaphase chromosome. accumulates into the central axis of chromatids from prometaphase through metaphase. Notably, the relo- nuclear throughout the cell cycle in S. cerevisiae calization of SMC2 coincides with NEBD and the [30,31]; in the yeast Schizosaccharomyces recruitment of condensin I to chromosomes. These pombe, however, most condensin molecules are cyto- data suggest that structural changes of plasmic during interphase, and transported into the chromosomes observed before and after NEBD may nucleus during mitosis in a Cdk1-dependent manner be mechanistically distinct (Figure 3C). Condensin II [32]. In vertebrate cells, condensin II is predominantly could initiate the early stage of condensation by hier- nuclear, whereas condensin I is sequestered in the archical folding. Upon NEBD, condensin I cooperates cytoplasm during interphase (Figure 3A) [33,34]. Con- with condensin II to shape, resolve and stabilize chro- densin I gains access to the chromosomes only after mosomes by forming an ‘axial glue’ structure the nuclear envelope breaks down in prometaphase, within the chromatids. and the two complexes alternate along the chromatid Maeshima and Laemmli [39] propose a different axis by metaphase [3,33]. A simple prediction from this model in which an early stage of condensation is observation is that an early stage of chromosome con- driven by II-mediated axis formation densation within the prophase nucleus may primarily and is followed by the action of condensin subunits. be mediated by condensin II. Small-interfering RNA This model does not, however, take into account the (siRNA) experiments in HeLa cells indeed support this role of condensin II, which was discovered only notion: depletion of condensin II-specific subunits recently. In the future, real-time analyses of distribu- delays prophase condensation, whereas depletion of tion and dynamics of these chromosomal components condensin I-specific subunits does not [33,34]. Con- will help test and refine the currently existing models. sistently, depletion of an SMC subunit also affects The observations described above raise an prophase condensation in chicken DT40 cells [35]. interesting question about the ‘reversibility’ of chro- However, such a clear division of labor between the mosome condensation. In response to various types two condensin complexes may not be applicable to all of stress, for example, as a result of chromosome metazoans. For instance, in Drosophila, which appar- damage or disassembly, cells in early ently has no gene for the CAP-G2 subunit, in prophase decondense their chromosomes and return the CAP-G subunit affect interphase gene to G2 phase, whereas such reversal of condensation expression [36,37]. does not occur in cells after late prophase [40]. How is the action of the two condensin complexes Recent studies [41,42] have suggested that this regulated during mitosis? In Xenopus egg extracts, ‘antephase’ checkpoint, activation of which requires Cdk1-dependent is at the of the p38 MAP kinase, monitors global chromatin topol- condensin regulation [2,27]. In somatic cells, two ogy before making the final commitment to mitosis major cyclin–Cdk complexes are known to regulate (Figure 3D). It seems plausible that the reversible Current Biology R269

Figure 3. Cell cycle regulation of NEBD condensins I and II. (A) The ‘traditional’ classification of A G2 pro prometa meta ana telo mitosis and subcellular distributions of condensins I and II during the cell cycle. Nuclear envelope breakdown (NEBD), Condensin I Nucleus one of the key events in early mitosis, occurs at the transition from prophase Cytoplasm (pro) to prometaphase (prometa) in this classification. The distributions of con- Condensin II densin I and condensin II complexes are shown in blue and magenta, respec- tively. (B) Sequential activation of cyclin B A–Cdk1 and cyclin B–Cdk1 during Cdk Cyclin A-cdk1 Cyclin B-cdk1 mitosis. (C) Hypothesized activities of activities condensins I and II during mitosis. According to this model, condensin II is C responsible for prophase condensation, Condensin Condensin I which may involve reversible, hierarchi- activities Condensin II cal folding. Upon NEBD, condensin I gains access to chromosomes and Hierarchical folding Axis formation cooperates with condensin II to build Reversible Irreversible fully resolved sister chromatids. This D 1234 5 process accompanies the formation of the chromatid axis and cannot be Commitment reversed by DNA damage. (D) A classifi- to mitosis cation of mitotic stages proposed by Current Biology Pines and Rieder [85], which empha- sizes the ‘point of no return’ that makes the final commitment to mitosis. phase of condensation might correspond to the stage recruiting condensins to chromosomes, available lines of condensin II-mediated hierarchical folding, and that of evidence do not support this notion [45,46]. A recent condensin II may be an integral component of this paper [47] reports that a subfraction of condensin sub- surveillance mechanism. For instance, condensin II- units co-purifies with the DNA mediated conformational changes of chromosomes DNMT3B, which further interacts with a might be used as a strategy to monitor potential deacetylase (HDAC1). Although the functional rele- chromosome aberrations. If such aberrations are vance of these interactions remains to be determined, found, the checkpoint is activated and reverses the exploring potential between condensins and action of condensin II possibly by down-regulating the epigenetic machineries will be an exciting future cyclin A–Cdk1 [43]. This checkpoint control is likely to direction in the field. be less stringent or even absent in early embryonic cells. Consistent with this notion, condensin II is much Condensins and Metaphase Chromosome less abundant, and its contribution to chromosome Architecture architecture is less prominent in early embryonic While early studies in Xenopus cell-free extracts [1,2] extracts than in somatic cells [3]. and yeast genetics [30,32,48–51] demonstrated that In metaphase chromosomes, condensins I and II condensin function is required for the establishment accumulate along the axis of sister chromatids, and maintenance of metaphase chromosome struc- possibly in an alternative fashion [3,33]. Biochemical ture, it remains controversial exactly how condensins experiments have shown that at least condensin I con- contribute to these processes [52,53]. For example, stitutes part of a fraction known as the chromosome individual masses of chromatin with a certain degree scaffold [35,39,44]. The two complexes can be tar- of compaction are observed in condensin mutants of geted to chromosomes independently of each other in Drosophila [36,54,55] and C. elegans [18] or in verte- both HeLa cells and Xenopus egg extracts [3,34]. brate cells depleted of condensin subunits [34,35], What, then, determines the differential distribution of leading to the suggestion that condensins may not be condensins I and II along the chromatids? It is possi- the sole factor required for chromatin compaction ble, but unlikely, that their distribution depends solely during mitosis. In this section, an attempt is made to on underlying DNA sequences. Given the observation clarify and discuss problems and difficulties that the that the relative abundance of condensins I and II is current field is faced with. different between the embryonic and somatic cells [3], First of all, it is clear that part of the controversy an epigenetic mechanism may exist that flexibly and stems from our lack of knowledge about how sister dynamically regulates the distribution of the two con- chromatid fibers are folded and organized to densin complexes along the arm. Currently, no specific assemble a metaphase chromosome [56]. This histone modification is known that affects targeting or problem leaves a large window for disparate interpre- function of condensins. While it has long been sus- tations of seemingly similar observations, and pro- pected that mitosis-specific phosphorylation of serine vides an opportunity to draw unconvincing 10 (and/or serine 28) of histone H3 might participate in conclusions from a limited amount of data. Figure 4 Review R270

Figure 4. Different types of chromo- ABC some ‘compaction’. (A–C) Three examples of hypothetical chromosome structure assembled Top view from a fixed volume of chromatin are viewed from top (first row) or side (second row). Each structure contains duplicated sister chromatids (shown in blue and magenta), the folding paths of which are shown in the third row. Side view Without any active folding mecha- Axial D nisms, the two chromatids would be lengthening converted into unresolved, random coils (A). One mechanism of ordered folding would produce a rod-shaped structure in which the two chromatids are folded together without being Chromatin resolved from each other (B). An addi- path tional mechanism would be required to make a ‘functional’ metaphase chro- mosome in which the two sister chro- Axial E Random coil matids are fully resolved and folded Ordered shortening separately (C). (D,E) Two representa- tive variations of the resolved chromo- Unresolved Resolved some with their volumes fixed. Axial Current Biology lengthening would create a longer and thinner chromosome (D), whereas axial shortening would produce a shorter and thicker chromosome (E). depicts three examples of hypothetical structures that chromatid resolution [54,55], but it remains to be fully can be produced from a fixed volume of chromatin. It established to what extent and how condensins is very important to distinguish between different participate in the ordered folding process. structures that may sometimes be considered equally The second problem, closely related to the first one, ‘compacted’. During mitosis, disassembly of the is the lack of reliable, quantitative assays to probe nuclear envelope allows release of chromatin fibers metaphase chromosome structure. It is well known from specific tethering within the interphase nucleus. that chromosome morphology is highly sensitive to a Without an active folding mechanism, this would number of chemical and physical treatments, and produce random coils (Figure 4A) rather than varies depending on different fixation and imaging extended chromatin fibers in the cell. Electrostatic techniques. At the same time, it is difficult to analyze interactions within the chromatin fibers and/or passive chromosome structure in great detail without aid of exclusion from the molecularly crowded cytoplasm some spreading techniques. In fact, the contribution may confer an additional level of compaction. This of condensins to metaphase chromosome architec- random compaction is clearly distinct from the con- ture can be demonstrated most convincingly when densation of mitotic chromosomes, which is a deter- chromosomes are subjected to hypotonic treatments ministic, shaping process rather than a simple linear [3,34,35]. Under such conditions, Ono et al. [3] compaction process [57,58]. showed that condensins I and II have distinct roles in It is also important to note that one of the chief determining the shape of metaphase chromosomes. functions of metaphase chromosome assembly is to On the other hand, Hudson et al. [35] devised a novel ‘resolve’ sister chromatids, an essential prerequisite assay in which chromosomes were exposed alter- for their rapid and synchronous separation at nately to a buffer containing MgCl2 or its chelator, and anaphase. In principle, one type of ordered folding demonstrated that the structural integrity of chromo- can produce rod-shaped chromosomes in which somes is severely compromised in the absence of juxtaposed sister chromatids are folded together SMC2. These results clearly underscore the funda- (Figure 4B), but these unresolved chromosomes are mental importance of condensins in shaping and unlikely to be segregated properly in anaphase. Only maintaining metaphase chromosome architecture. when fully resolved sister chromatids are constructed, The current approaches should, however, be comple- does the chromosomes become ‘functional’ — that is, mented by more quantitative and less invasive competent for anaphase segregation (Figure 4C). approaches in the future. Those may include live-cell Further variations of this structure include axial imaging analyses in which the density/volume of chro- lengthening and shortening, which accompany lateral matin is measured throughout the cell cycle or in contraction and expansion, respectively (Figure 4D,E). which the dynamics of specifically labeled chromoso- Thus, without knowing the actual folding path of the mal loci is followed in real time. chromatid fibers within a chromosome, it is often Finally, it should be emphasized that Xenopus cell- misleading to judge whether its assembly is normal or free extracts remain a very powerful experimental not. Currently, there is a clear consensus that system for studying mitotic chromosome architecture condensin function is essential for facilitating sister and dynamics. In general, much severer Current Biology R271

Figure 5. Multilayered contribution of condensins to mitotic chromosome segregation. Chromosome (A) Condensins play critical roles in the A bridges resolution of sister chromatids in Sister metaphase. Defects in this process chromatid lead to the formation of chromosome resolution bridges in the subsequent anaphase. Kinetochore B (B) Condensins regulate proper assem- orientation bly of centromeric and Abnormal thereby contribute to determining the microtubule back-to-back orientation of sister kine- attachment tochores (yellow). When this process is rDNA C compromised, abnormal interactions organization between kinetochores and micro- tubules are observed. (C) Condensins rDNA segregation may have a specialized role in the orga- defects nization and segregation of repetitive DNAs. Condensin’s participation in the Current Biology segregation of rDNA (orange) has been described in S. cerevisiae. are observed in extracts immunochemically depleted of defect is observed in many different organisms, condensins than in somatic tissue culture cells treated including yeast [48,49], Drosophila [55,63] and with condensin siRNAs. When both condensins I and II chicken DT40 cells [35], and is likely to be a direct are depleted from the extracts, no individual chromo- consequence of poor resolution and/or abnormal somes are assembled, regardless of the presence or compaction of sister chromatids in the preceding absence of prior DNA replication [2,59]. It has also been metaphase. In these cells, the kinetochores appear to shown that single depletion of condensin I or II pro- function normally and attempt to pull unresolved duces a drastically distinct [3]. One poten- chromatids to opposite poles without success (Figure tial explanation for these ‘clean’ phenotypes may be 5A). In Xenopus egg extracts [59] or human tissue that whole condensin complexes, either condensin I or culture cells [33], however, severe defects in kineto- II or both, are depleted from Xenopus -free chore-microtubule interactions, indicative of extracts. In contrast, only a single subunit (or at most merotelic attachment, are frequently observed after two subunits) is depleted by RNA interference or by condensin depletion (Figure 5B). Merotelic attach- conditional knockdown in tissue culture experiments. ment does not activate the spindle checkpoint [64], The fate of other non-target subunits is not fully char- consistent with the observation that condensin deple- acterized in the latter studies, with an assumption that tion does not induce robust arrest at mitosis. depletion of one subunit compromises all functions Abnormal attachment of kinetochore microtubules executed by the whole complex. A difference in the is even more prominently observed in condensin- efficiency of depletion may also lead to variable sever- deficient embryos of C. elegans [17,18]. In holocentric ity of defective phenotypes, as exemplified in recent chromosomes of C. elegans, numerous kinetochores studies in C. elegans [18,60]. Another explanation is assemble along the entire length of each chromatid, that the two experimental systems use different start- forming two ‘lines’ on the outer surfaces of a ing materials for chromosome assembly, which may in metaphase chromosome. Condensin II localizes turn produce seemingly different terminal phenotypes, underneath these structures (Figure 2C), and does so as discussed elsewhere [52]. Furthermore, the possibil- in an Aurora B-dependent manner. At first glance, this ity cannot be ruled out that chromosome assembly in distribution of condensin II in the C. elegans chromo- embryonic cells depends solely on condensins, somes is very different from that in vertebrate whereas somatic cells use an additional factor(s) chromosomes. A recent study [33], however, shows besides condensins. In this sense, it is intriguing to note that, in human cells, a subpopulation of condensin II is that even the relative abundance and functional contri- enriched at or near the inner kinetochore plate (Figure butions of condensins I and II are different between 2B), and that the kinetochore-specific localization of embryonic and somatic cells [3]. Moreover, measure- condensin II, but not its distribution along the arm, is ments of the mechanical properties of chromosomes — under the control of Aurora B. Thus, monocentric and for example, the stiffness and elasticity — detect differ- holocentric chromosomes do share many properties ences between embryonic and somatic ones [61,62]. in common. Condensins are likely to contribute to Thus, developmental regulation of chromosome archi- proper assembly of centromeric chromatin in both tecture is another interesting subject of future research. types of chromosomes, which in turn plays a critical role in establishing the back-to-back orientation of Contributions of Condensins to Specific sister kinetochores. Chromosomal Domains Kinetochore orientation rDNA segregation One of the classical phenotypes observed in A number of recent papers [65–68] report that condensin-deficient cells is a massive amount of condensin’s recruitment and function are regulated by chromosome bridges in anaphase. This segregation a unique mechanism at the rDNA locus in S. cerevisiae. Review R272

While most chromosomal regions are segregated upon required for meiotic (but not mitotic) chromosome of a cohesin subunit at the segregation [71], providing an additional level of metaphase/anaphase transition, rDNA segregation complexity to this problem. In vertebrates and plants, occurs in mid-anaphase and requires a pathway it will be of great importance to determine whether involving Cdc14, a protein phosphatase that is acti- condensins I and II are subjected to different, tempo- vated by the ‘fourteen early anaphase release’ (FEAR) ral and spatial regulation during meiosis, and whether network. Condensin is recruited to the rDNA locus in they play non-overlapping functions in meiotic anaphase in a Cdc14-dependent manner and mediates chromosome recombination and segregation. rDNA condensation and resolution (Figure 5C). As has been demonstrated in previous studies of Ipl1/Aurora kinase activity is required for the conden- cohesin [15], further characterization of condensins sation process, but not for resolution. during meiosis should not only facilitate our under- The molecular nature of the cohesin-independent standing of their meiosis-specific functions but also linkage that persists until mid-anaphase at the rDNA provide deeper insights into their basic functions locus is unknown. The primary candidate for such a during mitosis. It will be of particular interest to eluci- linkage is the catenation between sister chromatids, date the mechanism of condensin’s action in resolv- resolution of which requires a combined action of ing the recombination-dependent linkages of meiosis condensin and topoisomerase II (although one study I chromosomes [69], which may also occur during argues against this possibility [67]). Alternatively, con- mitosis at repetitive loci such as rDNA and densin function may be required to resolve inter- [67,72]. By analogy to cohesin [73–75], condensins repeat recombination in the rDNA array that would could also use a unique set of subunits to execute otherwise impede segregation [31]. Interestingly, yeast their specialized functions in meiotic cells. Although condensin subunits are subjected to multiple post- no such candidates have been found so far by bioin- translational modifications in anaphase. For example, formatic approaches, this may not be surprising the Ycg1/CAP-G subunit of condensin is phosphory- because even the similarity between the non-SMC lated in an Ipl1/Aurora-dependent manner [65], and subunits of condensins I and II is very limited [3]. A the Ycs4/CAP-D2 subunit is sumoylated in a Cdc14- combination of genetics and may yet stimulated manner [67]. The functional significance of identify meiosis-specific condensin subunits. these modifications remains to be determined, however. The repetitive nature of rDNA in yeast may Condensins, Gene Regulation and Genome Stability offer an excellent model system for studying the Accumulating lines of evidence suggest that action of condensins in organisms with more complex condensins have important functions in chromosome . regulation outside mitosis and meiosis. In S. cere- visiae, transcriptional silencing is altered in a locus- Condensins and Meiotic Chromosome Functions specific manner in condensin mutants [31]. For Given the fundamental functions of condensins in example, silencing is enhanced at the rDNA locus but mitotic chromosome assembly and segregation, it is reduced at telomeres, and this change is accompa- not surprising to find that condensins also play nied by relocalization of the silencing factor Sir2 from crucial roles in the structural and functional organiza- telomeres to the rDNA arrays [76]. In S. pombe, a tion of meiotic chromosomes. In S. cerevisiae, con- condensin mutant displays hypersensitivity to a treat- densin subunits localize to the axial core of ment that slows down DNA replication and fails to pachytene chromosomes and contribute to their axial activate the checkpoint kinase Cds1/Chk2 under such compaction and individualization [69]. Chromosomal a condition [77]. In Drosophila, condensin function is localization of Zip1, a component of the central required for transcriptional repression in centromere- element, is severely disturbed in condensin mutants, proximal heterochromatin [36], and its resulting in improper assembly of the synaptonemal exacerbates the ‘rough eye’ phenotype that is induced complex. As a consequence, homologue pairing and by overexpression of the centromere-specific histone processing of double-strand breaks are perturbed in variant CENP-A [37]. Although global defects in chro- these mutant cells. Evidence is also available that matin structure may be sufficient to account for these condensin is required for the resolution of recombi- diverse phenotypes observed in the condensin nation-dependent linkages between homologues in mutants, more specific involvement of condensin sub- meiosis I and perhaps for the segregation of sister units in each event cannot be excluded. In verte- chromatids in meiosis II as well. brates, potential contribution of condensin II to the A requirement for condensin function in both structural and functional organization of interphase meiosis I and meiosis II is consistent with results from chromatin remains to be explored. Arabidopsis [70] and C. elegans [60]. Unlike in S. cere- An elegant series of genetic and biochemical visiae, in C. elegans the condensin subunits associate studies [19] has revealed that a condensin-like with chromosomes only after the exit from pachytene, complex regulates dosage compensation in C. and restructure them in preparation for the subse- elegans. The dosage compensation complex (DCC) is quent two meiotic divisions. The difference may be likely to be the outcome of an evolutionary explained by the fact that S. cerevisiae has condensin that arose to mediate chromosome-wide gene repres- I alone, whereas C. elegans contains condensin II sion in the nematode. A crucial question is how the only. However, the non-SMC components of the DCC is specifically targeted to both X chromosomes dosage compensation complex in C. elegans are also in XX animals and reduces the level of transcripts by Current Biology R273

half. Recent work [78] has identified cis-acting DNA address the evolution and developmental regulation of elements that recruit this complex to the X chromo- chromosome architecture from a completely new per- some and facilitate its spreading along the entire spective. It is anticipated that the structural and func- length of the chromosome. Intriguingly, the same tional characterization of condensins will continue to complex is also targeted to the sex-determining auto- provide new and surprising insights into chromosome somal gene her-1 and represses its >20- biology and beyond. fold [79]. It is not known how the DCC is able to support the two different modes of gene repression: Acknowledgments the two-fold chromosome-wide repression and I thank members of the Hirano laboratory for critically reading twenty-fold gene-specific repression. the manuscript. I am also grateful to many colleagues in the Might any of the non-mitotic functions of con- field for stimulating discussions. The work from the author’s densins be linked to human diseases in the future? laboratory was supported by grants from the National Given their fundamental functions in chromosome Institutes of Health. segregation, it is not surprising to find that serendipi- References tous knocking-out of the gene encoding the CAP-G2 1. Hirano, T., and Mitchison, T.J. (1994). A heterodimeric coiled-coil subunit caused embryonic lethality in mice [80]. protein required for mitotic chromosome condensation in vitro. Cell Recent studies, however, show that haploinsufficiency 79, 449–458. 2. Hirano, T., Kobayashi, R., and Hirano, M. (1997). Condensins, chro- of a cohesin regulator (NIPBL/Nipped-B/Scc2) causes mosome condensation protein complexes containing XCAP-C, Cornelia de Lange syndrome, a human disease char- XCAP-E and a Xenopus homolog of the Drosophila Barren protein. acterized by multiple developmental defects [81,82]. Cell 89, 511–521. 3. Ono, T., Losada, A., Hirano, M., Myers, M.P., Neuwald, A.F., and Cases may therefore be found in which hypomorphic Hirano, T. (2003). Differential contributions of condensin I and con- mutations in condensin subunits (or their regulators) densin II to mitotic chromosome architecture in vertebrate cells. causes specific developmental defects in . Cell 115, 109–121. 4. Yeong, F.M., Hombauer, H., Wendt, K.S., Hirota, T., Mudrak, I., One intriguing example is premature chromosome Mechtler, K., Loregger, T., Marchler-Bauer, A., Tanaka, K., Peters, condensation (PCC) syndrome in humans, where the J.-M., et al. (2003). Identification of a subunit of a novel kleisin- β/SMC complex as a potential substrate of protein phosphatase 2A. gene responsible was found to be allelic to one of the Curr. Biol. 13, 2058–2064. genes mutated in microcephaly, an autosomal devel- 5. Melby, T.E.G., Ciampaglio, C.N., Briscoe, G., and Erickson, H.P. opment disorder characterized by a marked reduction (1998). The symmetrical structure of structural maintenance of chro- mosomes (SMC) and MukB proteins: long, antiparallel coiled coils, in brain size [83]. In mutant cells derived from PCC folded at a flexible hinge. J. Cell Biol. 142, 1595–1604. patients, a large fraction of G2 cells displays an abnor- 6. Anderson, D.E., Losada, A., Erickson, H.P., and Hirano, T. (2002). mal ‘prophase-like’ morphology of chromosomes, Condensin and cohesin display different arm conformations with characteristic hinge angles. J. Cell Biol. 156, 419–424. although subsequent segregation is apparently 7. Lammens, A., Schele, A., and Hopfner, K.-P. (2004). Structural bio- normal. It will be of interest to determine whether con- chemistry of ATP-driven dimerization and DNA-stimulated activa- densin function may be prematurely activated in these tion of SMC ATPases. Curr. Biol. 14, 1778–1782. mutant cells. 8. Haering, C.H., Schoffegger, D., Nishino, T., Helmhart, W., Nasmyth, K., and Lowe, J. (2004). Structure and stability of cohesin’s Smc1- kleisin interaction. Mol. Cell 15, 951–964. Future Directions 9. Hirano, M., Anderson, D.E., Erickson, H.P., and Hirano, T. (2001). Bimodal activation of SMC ATPase by intra- and inter-molecular Since the initial discovery of their subunits a decade interactions. EMBO J. 20, 3238–3250. ago, condensin complexes have gained much atten- 10. Hirano, M., and Hirano, T. (2004). Positive and negative regulation tion in the field. Although the characterization of con- of SMC-DNA interactions by ATP and accessory proteins. EMBO J. densins through a wide variety of approaches has 23, 2664–2673. 11. Neuwald, A.F., and Hirano, T. (2000). HEAT repeats associated with greatly enriched our understanding of chromosome condensins, and other chromosome-related complexes. architecture and dynamics, a number of fundamental Genome Res. 10, 1445–1452. questions remain to be answered. One of the important 12. Schleiffer, A., Kaitna, S., Maurer-Stroh, S., Glotzer, M., Nasmyth, K., and Eisenhaber, F. (2003). Kleisins: a superfamily of bacterial and goals is to fully elucidate the molecular action of con- eukaryotic SMC protein partners. Mol. Cell 11, 571–575. densins. How does the two-armed structure of con- 13. Yoshimura, S.H., Hizume, K., Murakami, A., Sutani, T., Takeyasu, K., densins support their dynamic interactions with DNA and Yanagida, M. (2002). Condensin architecture and interaction with DNA: regulatory non-SMC subunits bind to the head of SMC and chromatin fibers in vitro? How different is the heterodimer. Curr. Biol. 12, 508–513. action of condensins from that of cohesin [84]? Such 14. Kimura, K., and Hirano, T. (2000). Dual roles of the 11S regulatory information will be essential to fill the gap that currently subcomplex in condensin functions. Proc. Natl. Acad. Sci. USA 97, 11972–11977. exists between the biochemistry and of 15. Nasmyth, K. (2001). Disseminating the genome: joining, resolving, condensins. Mechanistically, how do condensins par- and separating sister chromatids during mitosis and meiosis. Annu. ticipate in determining and maintaining the shape of Rev. Genet. 35, 673–745. 16. Matsuzaki, M., Misumi, O., Shin-I, T., Maruyama, S., Takahara, M., metaphase chromosomes? Do any other factors Miyagishima, S.Y., Mori, T., Nishida, K., Yagisawa, F., Nishida, K., et essential for this process remain to be discovered? al. (2004). Genome sequence of the ultrasmall unicellular red alga It has become increasingly clear that the Cyanidioschyzon merolae 10D. Nature 428, 653–657. 17. Stear, J.H., and Roth, M.B. (2002). Characterization of HCP-6, a C. fundamental roles of condensins in chromosome elegans protein required to prevent chromosome twisting and organization and dynamics range far beyond the con- merotelic attachment. Genes Dev. 16, 1498–1508. densation of mitotic chromosomes. Perhaps one of 18. Hagstrom, K.A., Holmes, V.F., Cozzarelli, N.R., and Meyer, B.J. (2002). C. elegans condensin promotes mitotic chromosome archi- the most exciting areas in the future is to address tecture, centromere organization, and sister chromatid segregation potential involvement of condensins in epigenetic during mitosis and meiosis. Genes Dev. 16, 729–742. phenomena. Moreover, the identification of condensin 19. Hagstrom, K.A., and Meyer, B.J. (2003). Condensin and cohesin: more than chromosome compactor and glue. Nat. Rev. Genetics 4, II has provided us with an excellent opportunity to 520–534. Review R274

20. Volkov, A., Mascarenhas, J., Andrei-Selmer, C., Ulrich, H.D., and 44. Saitoh, N., Goldberg, I.G., Wood, E.R., and Earnshaw, W.C. (1994). Graumann, P.L. (2003). A prokaryotic condensin/cohesin-like ScII: an abundant chromosome is a member of a complex can actively compact chromosomes from a single position family of putative ATPases with an unusual predicted tertiary struc- on the and binds to DNA as a ring-like structure. Mol. Cell. ture. J. Cell Biol. 127, 303–318. Biol. 23, 5638–5650. 45. MacCallum, D.E., Losada, A., Kobayashi, R., and Hirano, T. (2002). 21. Sutani, T., and Yanagida, M. (1997). DNA renaturation activity of the ISWI remodeling complexes in Xenopus egg extracts: identification SMC complex implicated in chromosome condensation. Nature as major chromosomal components that are regulated by INCENP- 388, 798–801. aurora B. Mol. Biol. Cell 13, 25–39. 22. Sakai, A., Hizume, K., Sutani, T., Takeyasu, K., and Yanagida, M. 46. Murnion, M.E., Adams, R.R., Callister, D.M., Allis, C.D., Earnshaw, (2003). Condensin but not cohesin SMC heterodimer induces DNA W.C., and Swedlow, J.R. (2001). Chromatin-associated protein reannealing through protein-protein assembly. EMBO J. 22, phosphatase regulates aurora-B and histone H3 phosphorylation. 2764–2775. J. Biol. Chem. 276, 26656–26665. 23. Stray, J.E., and Lindsley, J.E. (2003). Biochemical analysis of the 47. Geiman, T.M., Sankpal, U.T., Robertson, A.K., Chen, Y., Mazumdar, yeast condensin Smc2/4 complex: an ATPase that promotes knot- M., Heale, J.T., Schmiesing, J.A., Kim, W., Yokomori, K., Zhao, Y., ting of circular DNA. J. Biol. Chem. 278, 26238–26248. et al. (2004). Isolation and characterization of a novel DNA methyl- transferase complex linking DNMT3B with components of the 24. Kimura, K., and Hirano, T. (1997). ATP-dependent positive super- mitotic chromosome condensation machinery. Nucleic Acids Res. coiling of DNA by 13S condensin: a biochemical implication for 32, 2716–2729. chromosome condensation. Cell 90, 625–634. 48. Saka, Y., Sutani, T., Yamashita, Y., Saitoh, S., Takeuchi, M., 25. Kimura, K., Rybenkov, V.V., Crisona, N.J., Hirano, T., and Cozzarelli, Nakaseko, Y., and Yanagida, M. (1994). Fission yeast cut3 and N.R. (1999). 13S condensin actively reconfigures DNA by introduc- cut14, members of a ubiquitous protein family, are required for ing global positive writhe: implications for chromosome condensa- chromosome condensation and segregation in mitosis. EMBO J. tion. Cell 98, 239–248. 13, 4938–4952. 26. Bazett-Jones, D.P., Kimura, K., and Hirano, T. (2002). Efficient 49. Strunnikov, A.V., Hogan, E., and Koshland, D. (1995). SMC2, a Sac- supercoiling of DNA by a single condensin complex as revealed by charomyces cerevisiae gene essential for chromosome segregation electron spectroscopic imaging. Mol. Cell 9, 1183–1190. and condensation, defines a subgroup within the SMC family. 27. Kimura, K., Hirano, M., Kobayashi, R., and Hirano, T. (1998). Phos- Genes Dev. 9, 587–599. phorylation and activation of 13S condensin by Cdc2 in vitro. 50. Lavoie, B.D., Tuffo, K.M., Oh, S., Koshland, D., and Holm, C. (2000). Science 282, 487–490. Mitotic chromosome condensation requires Brn1p, the yeast 28. Strick, T.R., Kawaguchi, T., and Hirano, T. (2004). Real-time detec- homolog of Barren. Mol. Biol. Cell 11, 1293–1304. tion of single-molecule DNA compaction by condensin I. Curr. Biol. 51. Lavoie, B.D., Hogan, E., and Koshland, D. (2002). In vivo dissection 14, 874–880. of the chromosome condensation machinery: reversibility of con- 29. Case, R.B., Chang, Y.P., Smith, S.B., Gore, J., Cozzarelli, N.R., and densation distinguishes contributions of condensin and cohesin. J. Bustamante, C. (2004). The bacterial condensin MukBEF compacts Cell Biol. 156, 805–815. DNA into a repetitive, stable structure. Science 305, 222–227. 52. Hirano, T. (2004). Chromosome shaping by two condensins. Cell 30. Freeman, L., Aragon-Alcaide, L., and Strunnikov, A.V. (2000). The Cycle 3, 26–28. condensin complex governs chromosome condensation and 53. Gassmann, R., Vagnarelli, P., Hudson, D., and Earnshaw, W.C. mitotic transmission of rDNA. J. Cell Biol. 149, 811–824. (2004). Mitotic chromosome formation and the condensin paradox. 31. Bhalla, N., Biggins, S., and Murray, A.W. (2002). Mutation of YCS4, Exp. Cell Res. 296, 35–42. a budding yeast condensin subunit, affects mitotic and nonmitotic 54. Steffensen, S., Coelho, P.A., Cobbe, N., Vass, S., Costa, M., Hassan, chromosome behavior. Mol. Biol. Cell 13, 632–645. B., Prokopenko, S.N., Bellen, H., Heck, M.M.S., and Sunkel, C.E. 32. Sutani, T., Yuasa, T., Tomonaga, T., Dohmae, N., Takio, K., and (2001). A role for Drosophila SMC4 in the resolution of sister chro- Yanagida, M. (1999). Fission yeast condensin complex: essential matids in mitosis. Curr. Biol. 11, 295–307. roles of non-SMC subunits for condensation and cdc2 phosphory- 55. Coelho, P., Queiroz-Mechado, J., and Sunkel, C.E. (2003). Con- lation of Cut3/SMC4. Genes Dev. 13, 2271–2283. densin-dependent localisation of topoisomerase II to an axial chro- mosomal structure is required for sister chromatid resolution during 33. Ono, T., Fang, Y., Spector, D., and Hirano, T. (2004). Spatial and mitosis. J. Cell Sci. 116, 4763–4776. temporal regulation of condensins I and II in mitotic chromosome assembly in human cells. Mol. Biol. Cell 15, 3296–3308. 56. Swedlow, J.R., and Hirano, T. (2003). The making of the mitotic chromosome: modern insights into classical questions. Mol. Cell 11, 34. Hirota, T., Gerlich, D., Koch, B., Ellenberg, J., and Peters, J.M. 557–569. (2004). Distinct functions of condensin I and II in mitotic chromo- some assembly. J. Cell Sci. 117, 6435–6445. 57. Koshland, D., and Strunnikov, A. (1996). Mitotic chromosome con- densation. Annu. Rev. Cell Dev. Biol. 12, 305–333. 35. Hudson, D.F., Vagnarelli, P., Gassmann, R., and Earnshaw, W.C. (2003). Condensin is required for nonhistone protein assembly and 58. Hirano, T. (2000). Chromosome cohesion, condensation and sepa- structural integrity of vertebrate chromosomes. Dev. Cell 5, ration. Annu. Rev. Biochem. 69, 115–144. 323–336. 59. Wignall, S.M., Deehan, R., Maresca, T.J., and Heald, R. (2003). The 36. Dej, K.J., Ahn, C., and Orr-Weaver, T.L. (2004). Mutations in the condensin complex is required for proper spindle assembly and Drosophila condensin subunit dCAP-G: Defining the role of con- chromosome segregation in Xenopus egg extracts. J. Cell Biol. 161, densin for chromosome condensation in mitosis and gene expres- 1041–1051. sion in interphase. Genetics 168, 895–906. 60. Chan, R.C., Severson, A.F., and Meyer, B.J. (2004). Condensin 37. Jager, H., Rauch, M., and Heidmann, S. (2005). The Drosophila restructures chromosomes in preparation for meiotic divisions. J. melanogaster condensin subunit Cap-G interacts with the cen- Cell Biol. 167, 613–625. tromere-specific histone H3 variant CID. Chromosoma 113, 61. Almagro, S., Riveline, D., Hirano, T., Houchmandzadeh, B., and 350–361. Dimitrov, S. (2004). The mitotic chromosome: an assembly of rigid elastic axes, organized by SMC proteins and surrounded by a soft 38. Kireeva, N., Lakonishok, M., Kireev, I., Hirano, T., and Belmont, A.S. chromatin envelope. J. Biol. Chem. 279, 5118–5126. (2004). Visualization of early chromosome condensation: a hierar- chical folding, axial glue model of chromosome structure. J. Cell 62. Poirier, M.G., Eroglu, S., and Marko, J.F. (2002). The bending rigid- Biol. 166, 775–785. ity of mitotic chromosomes. Mol. Biol. Cell 13, 2170–2179. 39. Maeshima, K., and Laemmli, U.K. (2003). A two-step scaffolding 63. Bhat, M.A., Philp, A.V., Glover, D.M., and Bellen, H.J. (1996). Chro- model for mitotic chromosome assembly. Dev. Cell 4, 467–480. matid segregation at anaphase requires the barren product, a novel chromosome associated protein that interacts with topoisomerase 40. Rieder, C.L., and Cole, R.W. (1998). Entry into mitosis in vertebrate II. Cell 87, 1103–1114. somatic cells is guarded by a chromosome damage checkpoint that 64. Cimini, D., Howell, B., Maddox, P., Khodjakov, A., Degrassi, F., and reverses the cell cycle when triggered during early but not late , E.D. (2001). Merotelic kinetochore orientation is a major prophase. J. Cell Biol. 142, 1013–1022. mechanism of aneuploidy in mitotic mammalian tissue cells. J. Cell 41. Matsusaka, T., and Pines, J. (2004). Chfr acts with the p38 stress Biol. 153, 517–527. kinases to block entry to mitosis in mammalian cells. J. Cell Biol. 65. Lavoie, B.D., Hogan, E., and Koshland, D. (2004). In vivo require- 166, 507–516. ments for rDNA chromosome condensation reveal two cell-cycle- 42. Mikhailov, A., Shinohara, M., and Rieder, C.L. (2004). Topoiso- regulated pathways for mitotic chromosome folding. Genes Dev. merase II and inhibitors delay the G2/M transi- 18, 76–87. tion by triggering the p38 MAPK checkpoint pathway. J. Cell Biol. 66. Sullivan, M., Higuchi, T., Katis, V.L., and Uhlmann, F. (2004). Cdc14 166, 517–526. phosphatase induced rDNA condensation and resolves cohesin- 43. Furuno, N., den Elzen, N., and Pines, J. (1999). Human cyclin A is independent cohesion during budding yeast anaphase. Cell 117, required for mitosis until mid prophase. J. Cell Biol. 147, 295–306. 471–482. Current Biology R275

67. D’Amours, D., Stegmeier, F., and Amon, A. (2004). Cdc14 and con- densin controls the dissolution of cohesin-independent chromo- some linkages at repeated DNA. Cell 117, 455–469. 68. Wang, B.-D., Yong-Gonzalez, V., and Strunnikov, A.V. (2004). Cdc14p/FEAR pathway controls segregation of in S. cerevisiae by facilitating condensin targeting to rDNA chromatin in anaphase. Cell Cycle 3, 960–967. 69. Yu, H.-G., and Koshland, D.E. (2003). Meiotic condensin is required for proper chromosome compaction, SC assembly, and resolution of recombination-dependent chromosome linkage. J. Cell Biol. 163, 937–947. 70. Siddiqui, N.U., Stronghill, P.E., Dengler, R.E., Hasenkampf, C.,A., and Riggs, C.D. (2003). Mutations in Arabidopsis condensin genes disrupt embryogenesis, meristem organization and segregation of homologous chromosomes during meiosis. Development 130, 3283–3295. 71. Lieb, J.D., Capowski, E.E., Meneely, P., and Meyer, B.J. (1996). DPY-26, a link between dosage compensation and meiotic chro- mosome segregation in the nematode. Science 274, 1732–1736. 72. Dynek, J.N., and Smith, S. (2004). Resolution of sister association is required for progression through mitosis. Science 304, 97–100. 73. Molnar, M., Bahler, J., Sipiczki, M., and Kohli, J. (1995). The rec8 gene of Schizosaccharomyces pombe is involved in linear element formation, chromosome pairing and sister-chromatid cohesion during meiosis. Genetics 141, 61–73. 74. Prieto, I., Suja, J.A., Pezzi, N., Kremer, L., Martinez-A., C., Rufas, J.S., and Barbero, J.L. (2001). Mammalian STAG3 is a cohesin spe- cific to sister chromatid arms during meiosis I. Nat. Cell Biol. 3, 761–766. 75. Revenkova, E., Eijpe, M., Heyting, C., Hodges, C.A., Hunt, P.A., B., L., Scherthan, H., and Jessberger, R. (2004). Cohesin SMC1β is required for meiotic chromosome dynamics, sister chromatid cohe- sion and DNA recombination. Nat. Cell Biol. 6, 555–562. 76. Machin, F., Paschos, K., Jarmuz, A., Torres-Rosell, J., Pade, C., and Aragon, L. (2004). Condensin regulates rDNA silencing by modulat- ing nucleolar Sir2p. Curr. Biol. 14, 125–130. 77. Aono, N., Sutani, T., Tomonaga, T., Mochida, S., and Yanagida, M. (2002). Cnd2 has dual roles in mitotic condensation and interphase. Nature 417, 197–202. 78. Csankovszki, G., McDonel, P., and Meyer, B.J. (2004). Recruitment and spreading of the C. elegans dosage compensation complex along X chromosomes. Science 303, 1182–1185. 79. Chu, D.S., Dawes, H.E., Lieb, J.D., Chan, R.C., Kuo, A.F., and Meyer, B.J. (2002). A molecular link between gene-specific and chromo- some-wide transcriptional repression. Genes Dev. 16, 796–805. 80. Smith, E.D., Xu, Y., Tomson, B.N., Leung, C.G., Fujikawa, Y., Orkin, S.H., and Crispino, J.D. (2004). More than , a novel gene required for mammalian postimplantation development. Mol. Cell. Biol. 24, 1168–1173. 81. Krantz, I.D., McCallum, J., DeScipio, C., Kaur, M., Gillis, L.A., Yaeger, D., Jukofsky, L., Wasserman, N., Bottani, A., Morris, C.A., et al. (2004). Cornelia de Lange syndrome is caused by mutations in NIPBL, the human homolog of Nipped-B. Nat. Genet. 36, 631–635. 82. Tonkin, E.T., Wang, T.J., Lisgo, S., Bamshad, M.J., and Strachan, T. (2004). NIPBL, encoding a homolog of fungal Scc2-type sister chro- matid cohesion proteins and fly Nipped-B, is mutated in Cornelia de Lange syndrome. Nat. Genet. 36, 636–641. 83. Trimborn, M., Bell, S.M., Felix, C., Rashid, Y., Jafri, H., Griffith, P.D., Neumann, L.M., Krebs, A., Reis, A., Sperling, K., et al. (2004). Muta- tions in microcephalin cause aberrant regulation of chromosome condensation. Am. J. Hum. Genet. 75, 261–266. 84. Haering, C.H., and Nasmyth, K. (2003). Building and breaking bridges between sister chromatids. Bioessays 25, 1178–1191. 85. Pines, J., and Rieder, C.L. (2001). Re-staging mitosis: a contempo- rary view of mitotic progression. Nat. Cell Biol. 3, E3–E6.