REVIEWS

Coupling changes in cell shape to segregation

Nitya Ramkumar and Buzz Baum Abstract | Animal cells undergo dramatic changes in shape, mechanics and polarity as they progress through the different stages of cell division. These changes begin at mitotic entry, with cell–substrate adhesion remodelling, assembly of a cortical actomyosin network and osmotic swelling, which together enable cells to adopt a near spherical form even when growing in a crowded tissue environment. These shape changes, which probably aid spindle assembly and positioning, are then reversed at mitotic exit to restore the cell morphology. Here, we discuss the dynamics, regulation and function of these processes, and how cell shape changes and sister chromatid segregation are coupled to ensure that the daughter cells generated through division receive their fair inheritance.

Astral microtubules During cell division, the entire set of cellular compo­ different systems remains to be determined, the spheri­ A subpopulation of dynamic nents must be segregated before being partitioned into cal shape is likely to provide a suitable environment for microtubules, which originate two daughter cells. This includes cellular constituents the spindle to assemble, while also delimiting the space from the centrosomes but do that are present in relatively large numbers, such as in which astral microtubules have to search to capture not attach to the 9 . They are ribosomes, which are partitioned by stochastic pro­ mitotic chromosomes . In addition, by simplifying cell present only during . cesses at division; cellular components that are pres­ geometry, mitotic rounding might contribute to the ent in smaller numbers, such as mitochondria and accurate partitioning of cellular contents into the two the Golgi, which tend to be fragmented and actively daughter cells. In a similar manner, the apical move­ transported to opposing cell poles to facilitate their ment of cell mass accompanying mitotic rounding in fair inheritance1; and structures that must be segre­ cells dividing in tall epithelia, such as neuroepithe­ gated with high precision, because their function is lia10, facilitates the preservation of apical polarity cues dependent on exact copy number, such as centrioles and cell–cell adhesion during a symmetrical division. and chromosomes. Therefore, although the basic goal Rounding also helps to set the stage for the dramatic of division is the same across all kingdoms of life2,3, it is redistribution of cell mass that is associated with especially challenging for eukaryotic cells, in which anaphase elongation and cytokinesis, which is impor­ segregation of genetic material must be ­coordinated tant for the restoration of normal cell packing in a in space and time with organelle segregation and cyto­ ­tissue that has been subject to mechanical stretching11. kinesis. The problem is further compounded for cells To aid division, changes in cell shape during mitotic in multicellular tissues such as epithelia, in which entry and exit are coupled to the clock by the cells are highly polarized in shape and structure and activity of the master mitotic kinase cyclin-dependent physically connected to one another. In the context kinase 1 (CDK1)–cyclin B. Cellular architecture begins of a tissue, cell divisions must also be resistant to the to be remodelled during prophase, as CDK1–cyclin B potentially adverse influence of extrinsic forces, while levels rise, causing cells to adopt a typical, spherical responding to cues that determine the axis of cell mitotic cell shape12, while the centrosomes separate division to facilitate the maintenance of polarity and and begin to nucleate microtubules to capture and tissue relaxation4,5. align chromosomes. These changes in cell shape are In animal cells, all of these processes are facilitated reversed following the drop in CDK1–cyclin B levels MRC Laboratory of Molecular Cell Biology, University by ‘mitotic rounding’. This is the process by which cells at anaphase. Then, as sister chromatids separate, cells College London, Gower Street, adopt a relatively simple, symmetric and spherical leaving mitosis elongate to make space for the extend­ London WC1E 6BT, UK. shape as they enter mitosis. Although this is not a uni­ ing spindle, while simultaneously assembling the con­ 6,7 Correspondence to B.B. versal phenomenon , most eukaryotic cells that lack tractile ring machinery to generate two daughter cells. [email protected] a rigid cell wall8, and therefore have a flexible form, The two processes must be highly coordinated in space doi:10.1038/nrm.2016.75 begin to ‘round up’ in this way as they enter mitosis. and time to ensure the equal inheritance of genetic Published online 29 June 2016 Although the precise function of mitotic rounding in material in the daughter cells.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 1 ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

In this Review, we discuss the dynamics of cell shape the phosphorylation of the proteome16–18. As a result, cell changes and their contribution to mitosis in cell cul­ shape and structure are rapidly altered within minutes ture and in the context of an epithelial tissue. Finally, of mitotic entry12. we explore the importance of crosstalk between the and the mitotic spindle for precise cell division. Adhesion remodelling during mitotic entry. One of the factors contributing to this rapid change in cell shape is Entry into mitosis loss of attachment to the underlying substrate. This loss For many animal cells, entry into mitosis begins with a of integrin-based adhesions is sufficient to cause cells in dramatic series of changes in cell shape, movement and culture to adopt a near-spherical shape — the minimum polarity10,13,14 (FIG. 1a,b). Although these changes have energy conformation for a cell with high cortical tension not been investigated in much detail, they are among (as the sphere has the least surface area for a given vol­ the first signs of a cell approaching mitosis. The changes ume). At a molecular level, this rapid change in adhesion are triggered by rising levels of CDK1–cyclin B15, along is probably triggered by activation of CDK1–cyclin B19–22. with rising activities of a cohort of mitotic kinases (most Although CDK1–cyclin B directly phosphorylates sev­ notably Aurora A, Aurora B and Polo-like kinase 1 eral adhesion complex components23, including integ­ (PLK1)). This sets up a positive feedback loop that rins24, focal adhesion kinase and paxillin25, the major culmin­ates in full kinase activation and a global shift in molecular event responsible for the loss of adhesions in

a NEB Anaphase ARP2/3 complex activity Isotrophic cortical network Contractile ring Actomyosin network

CDK1–cyclin B activity

Interphase Prophase Prometa Metaphase Anaphase

b Actin Chromatin Centrosome Furrow formation Microtubule Cell spreading

Substrate Adhesion Contractile Decreased actomyosin ring cell-substrate Cell division adhesion and movement

Cell rounding Polar relaxation

Assembly of cortical Cell elongation actomyosin network Figure 1 | Cell shape changes during mitosis. As animal cells become committed to mitosis, their internal architecture and physical connections to the environment are rapidly remodelled, causing cells to assumeNature a Reviewstypical, spherical | Molecular mitotic Cell cellBiology shape. These structural changes are reversed during mitotic exit as cells divide and revert to their initial interphase morphology. Structural changes are coupled to the cell cycle clock, and many are directed by changes in protein phosphorylation driven by the rise and fall of cyclin-dependent kinase 1 (CDK1)–cyclin B activity. a | As the levels of CDK1– cyclin B activity rise (blue line), the cell begins the assembly of its cortical actomyosin network (red line); both of these events are accelerated by loss of the nuclear–cytoplasmic compartment boundary. The cortical actomyosin network that is assembled upon entry into mitosis is largely independent of the activity of the actin nucleator ARP2/3 complex (purple line), which is inhibited at the onset of prophase. At anaphase, this mitotic actomyosin network is remodelled to generate a contractile actomyosin ring at the cell centre. The ARP2/3 complex is then reactivated as cells re-enter interphase. b | At the onset of mitosis, cells stop moving and reduce their adhesions to the substrate with rising levels of CDK1–cyclin B activity. Centrosomes separate, and cells begin to assemble a cortical actomyosin network and increase in height to round up. Rounding is expedited by nuclear envelope breakdown (NEB). Chromosomes (blue) then align at the metaphase plate. Once all the chromosomes are attached to the spindle and the spindle assembly checkpoint has been satisfied, the activation of the anaphase-promoting complex (APC) triggers the cleavage of cohesin, allowing sister chromatids to move to the opposite poles, and the degradation of cyclin B, leading to a collapse in CDK1 activity. This is accompanied by the relaxation of the cortical actomyosin network at cell poles and by the concentration of actomyosin at the cell centre to form a contractile ring. As cells divide, chromatids begin to de-condense, and the nuclear envelope and lamina reform.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrm ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

. Importantly, for cells in culture with Plasma pERM pSLIK membrane a isotropic interphase shape, it has also been suggested DIA that these retraction fibres provide cells with a physi­ DIA cal memory of the pattern of interphase cell–substrate adhesions, which then guides mitotic spindle orienta­ Actin tion27 (BOX 1) and the re‑establishment of these adhesions Myosin when cells re‑spread at mitotic exit28,29. Similar structures minifilament have been observed in mitotic cells within an epithe­ lium, which retain long basal retraction fibres as they Actin nucleation DIA RhoA ROK Myosin activity round up during mitosis. These fibres maintain physical ECT2 contact between cells and their underlying basal extra­ cellular matrix and seem to aid the restoration of apical basal polarity upon mitotic exit30,31. Importantly, epi­ adherens Actin thelial cells also retain their E‑cadherin-based Myosin ­junctions and tight junctions during the process of division pERM to preserve­ tissue integrity32,33. Chromosome Centrosome Assembling the mitotic cortical actin network. The adhesion remodelling that takes place as cells enter Microtubule mitosis is accompanied by profound changes in the Plasma membrane organization of cortical actin cytoskeleton (FIG. 2), which culminate in the assembly of a dense cortical actin net­ work character­istic of mitotic cells34. The loss of basal actin cables (also known as stress fibres) is likely to be a direct consequence of the disassembly of cell–substrate adhesions13. There is also a shift in the dominance of Substrate Focal adhesion different cortical actin nucleators at the onset of mitosis, Figure 2 | Cortical actomyosin network reorganization during mitotic rounding. from the ARP2/3 complex to formins14,35,36. For cells enter­ Nature Reviews | Molecular Cell Biology In isolated cells in culture, several factors contribute to the changes in the actomyosin ing mitosis in culture, this is apparent from the loss of network that aid in mitotic cell rounding. The activity of the actin nucleator ARP2/3 lamellipodia, which are actin-based membrane pro­ complex is reduced, whereas that of negative regulators of actin turnover is increased. trusions that are generated by branched actin filament Increasing levels of cyclin-dependent kinase 1 (CDK1)–cyclin B activity lead to nucleation downstream of Rac, the SCAR/WAVE com­ progressive loss of integrin-based cell–substrate adhesion. The rise in nuclear CDK1– plex37–39 and the ARP2/3 complex. Although the extent to cyclin B also leads to the export of ECT2 from the nucleus. This activates RhoA and its which residual ARP2/3 activity contributes to the gener­ targets, Diaphanous (DIA), Rho-associated protein kinase (ROK) and myosin (blue), 14,35,36 leading to the formation of a contractile actomyosin network. Following its mitotic ation of mitotic structures is currently unclear , this activation, the active phosphorylated SLIK (pSLIK) kinase (sterlie 20 kinase), now loss of lamellipodial actin is accompan­ied by the activ­ phosphorylates and activates ERM (Ezrin, Radixin, Moesin) proteins (Moesin in flies). ation of the formin-based actin nucleator Diaphanous Phosphorylated ERM (pERM; yellow) proteins tether this actomyosin network to the (DIA)14, which drives the assembly of a cortical net­

plasma membrane through binding transmembrane proteins and the phospholipid PIP2. work of actin filaments. Importantly, once activated, The cells retain actin-rich retraction fibres, which both direct spindle orientation (BOX 1) through physical association with Rho‑family GTPases, and help in cell re‑spreading following division. DIA gener­ates non-branched actin filaments. These ­provide an ideal substrate for non-muscle myosin II mini­ filaments40, which walk along antiparallel actin ­filaments many cell types seems to be inactivation of the RAP1 in a stepwise­ ­fashion to generate cortical tension. GTPase12. This is supported by the fact that many cul­ In human and fly cells, assembly of this DIA- tured cells expressing a constitutively activated form of dependent cortical actin mesh seems to be regulated RAP1 retain adhesions and remain flat as they enter by ECT2 (called Pbl in Drosophila melanogaster)13,14 mitosis, despite extensive CDK1–cyclin B-mediated — a potent Rho‑family GTPase activating protein41. phosphorylation of adhesion proteins12. This experiment ECT2 is released from the nucleus as levels of nuclear 13 Retraction fibres demonstrates the importance of adhesion remodelling CDK1–cyclin B increase at the onset of mitosis . Once Actin-rich fibres retained by for normal mitotic cell rounding. in the cytoplasm, ECT2 activates Rho‑family GTPases cells as they round up as they Although the loss of cell–substrate adhesions during at the plasma membrane (FIG. 2). These include RhoA42,43 enter mitosis. These hold the entry into mitosis is rapid, most animal cells maintain which, when bound to GTP, activates DIA and ROK cells in place and are thought to guide spindle orientation to physical connections with their environment through­ (Rho-associated protein kinase), which in turn activ­ 44 determine the division axis. out mitosis (FIG. 2). For animal cells in culture, the ates non-muscle myosin II . In addition, it has been actin-based structures that remain are called retraction suggested that ECT2 associates with the polarity pro­ Adherens junctions fibres26 (FIG. 2). Although these lack myosin II and do teins atypical protein kinase C (aPKC), partitioning Protein complexes consisting not provide robust cell–substrate adhesion (enabling defective 6 (PAR6) and CDC42, leading to the activ­ of cadherins and catenin, (REFS 14,45) which are involved in cell–cell mitotic cells to be dislodged by a physical blow to the ation of the Rho‑family GTPase CDC42 . junctions in epithelial and cell culture flask), it has been proposed that they act In this way, ECT2 may direct a shift in polarized actin endothelial cells. like tent cables5 — keeping rounded cells tethered to the organization, enabling epithelial cells to assemble an

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 3 ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

Box 1 | Spindle orientation guided by cortical cues in cellular biochemistry and cyto-architecture. This is augmented by osmotic swelling59 and a sudden influx Components of the molecular machinery that guide the orientation of the mitotic of water, leading to a significant (>10%) increase in cell spindle in response to cues in the cortex were first identified in genetic screens. These volume that is independent of the actin cytoskeleton60,61 include a small set of highly conserved proteins, G protein subunit G , LGN (also known αi and a dramatic increase in cell height59,62,63. In combin­ as GPSM2 or Pins) and nuclear mitotic apparatus protein 1 (NuMA; also known as Mud). Following nuclear envelope breakdown, these proteins localize to the cell cortex, ation, changes in cell architecture, actin remodelling, where they recruit the minus-end-directed motor dynein–dynactin146. As dynein walks cell–substrate adhesion and osmotic swelling, cause along astral microtubules, towards their minus ends and the spindle pole, it generates cells to assume a spherical shape by the time they have the forces required to reposition the spindle147. assembled a metaphase plate (FIGS 1,3a). Although many actin regulators (for example, afadin, myosin X, LIM kinase) have been implicated in spindle positioning, it is not clear whether individual actin regulators relay Changes in mechanical properties. Although spherical specific information to guide polarization of the cortex or whether they function mitotic cells resemble non-adherent interphase cells, collectively in construction of the mitotic cortical actin network. The latter is important, their mechanical properties are profoundly differ­ as the loss of cortical actin leads to the accumulation of cortical polarity cues together ent13,59,63–65. In fact, entry into mitosis is accompanied with dynein at centrosomes148. In this case, it is clear that actin is not needed for dynein by a rapid, nearly tenfold decrease in cell compliance to generate pulling forces but to provide a physical platform on the plasma membrane upon which polarity cues can be stably bound and read. This situation is further (equivalent to an increase in cell stiffness). This is complicated by the fact that spindle assembly depends on mitotic cell dependent on both osmotic swelling, which can be + + 59,60 geometry — which is another actin-dependent process. inhibited by the Na /H antiporter poison (EIPA) , and the cortical actomyosin network13,52. These forces are sufficient to deform neighbouring cells within a crowded epithelial tissue11,66 (FIG. 3b), can drive the api­ isotropic cortical actomyosin network as they round up cal movement of cell mass in a columnar epithelium63 to divide14. Interestingly, the same set of proteins drives and are strong enough to induce morphogenetic tissue both the assembly and the polarization of a cortical buckling in the developing fly and lumen growth in contractile actomyosin network before division of the zebrafish epithelial tissues66,67. Thus, the stable, rounded Caenorhabditis elegans zygote46. shape that is character­istic of metaphase cells reflects a Tight junctions (FIG. 3) Cell–cell junctions, consisting Many other changes in the dynamics and organiza­ balance of forces : the mitotic cortical actomyosin of occludins, claudins and tion of actin filaments contribute to the assembly and network, which is crosslinked to the plasma membrane associated proteins, which remodelling of the mitotic cortical actomyosin network. by active ERM proteins52, functions like the elastic skin bring membranes of adjacent For example, phosphorylation-induced changes in the of a ­balloon to counter the internal pressure established cells into close proximity to activities of proteins that negatively regulate actin fila­ by the activity of osmotically active ion pumps59,60. form a permeability barrier. They are positioned apical to ment formation, including LIM kinase, cofilin and WD the adherens junctions in repeat-containing protein 1 (WDR1), may contribute Mitotic exit vertebrates and (as septate to an increase in the actin filament turnover rate upon The duration of mitosis depends both on mitotic timers junctions) basal to adherens entry into mitosis35,47–50. Furthermore, the activation of and on satisfaction of the spindle assembly checkpoint68–71. junctions in most invertebrate epithelia. actin filament-binding proteins, such as ERM (Ezrin, When a minimal time has elapsed and the last pair of Radixin, Moesin) proteins, brought about by the SLIK chromosomes has been aligned at the metaphase plate ARP2/3 complex kinase51,52 leads to the physical crosslinking of the actin (reviewed in REF. 72), the anaphase-promoting complex A protein complex consisting filaments to proteins embedded in the plasma mem­ (APC; reviewed in REF. 73) is activated. This induces of seven polypeptides brane52,53 (FIG. 2). These changes in actin organization cleavage of cohesin, the protein complex that holds sis­ (including actin-related protein 2 (ARP2) and ARP3), are probably aided by a shift from the phospholipid ter chromatids together, enabling the movement of sister 54,55 which regulates the nucleation PIP3 towards PIP2 in the plasma membrane , which chromatids to opposing poles of the elongating anaphase of branched actin filament has been shown to activate ERM binding56. Together, spindle74 (FIG. 4a). At the same time, APC-mediated pro­ networks from the filament these changes in actin filament nucleation, turnover tein degradation reverses many of the regulatory changes minus end. and dynamics lead to the assembly of a thin but dense that accompany entry into mitosis, triggering the rapid Formins network of actin filaments and non-muscle myosin II degradation of cyclin B and the loss of mitotic kinase Proteins defined by the motors underneath the plasma membrane34. activity (reviewed in REF. 75). This drives cells out of presence of a formin Although this reorganization of the cytoskeleton mitosis — a process that culminates in cell division homology 2 (FH2) domain, begins in prophase, cytoskeletal remodelling acceler­ (reviewed in REF. 76). which nucleate actin filaments from growing (plus) ends to ates following the loss of the nuclear–cytoplasmic com­ generate parallel or antiparallel partment barrier (a process known as nuclear envelope Assembly and positioning of the actomyosin ring. filaments that are a good breakdown (NEB))57. One might speculate that this is Although many of the cell biological changes induced substrate for non-muscle due to the release of proteins previously sequestered in upon entry into mitosis are reversed during mitotic myosin II. the nucleus. In addition to residual nuclear ECT2, this exit (for example, reassembly of the nuclear compart­ Non-muscle myosin II includes RanGTP, which catalyses the release of pro­ ment; reviewed in REF. 77) the same is not true of actin An ATP-dependent motor teins from importins to alter microtubule dynamics and remodel­ling. In fact, the mitotic cortical actin network protein that forms large bipolar cortical actomyosin58. Simultaneously, the mixing of must not be disassembled at mitotic exit. This is because ‘minifilaments’. In non-muscle cytoplasm and nucleoplasm induced by the loss of the the machinery used to generate the actomyosin network cells, these both crosslink actin filaments and walk along nuclear compartment is likely to induce a sudden drop that establishes the characteristic spherical shape of the actin filaments to drive in the concentration of proteins localized to only one of metaphase cortex also controls the assembly of the acto- network contraction. the two compartments — inducing profound changes myosin contractile ring that is used to drive cytokinesis.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrm ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

Importins This includes ECT2, RhoA, DIA, actin, non-muscle formation, which divides the cell into two daughter cells 78 Protein complexes that myosin II and Anillin , all of which seem to be ­further (FIGS 1,4). The cortical remodelling that is triggered at transport proteins containing activated by the drop in CDK1–cyclin B activity at mitotic exit to facilitate these shape changes cannot nuclear localization sequences mitotic exit79,80. In this regard, mitotic rounding can be be subject to autonomous regulation; that is, an acto­ (NLS) into the nucleus. The complex consists of importin‑α seen as a prelude to division. Although the activity of this myosin ring cannot be simply assembled and constricted and importin‑β. set of actin regulators is required for the assembly of the at a fixed time and place following the activation of the actomyosin network that is necessary for both round­ APC. If each daughter cell is to inherit a single copy of Spindle assembly ing and cytokinesis, the assembly of an acto­myosin ring the nuclear genome, cells must ensure that the acto­ check point probably involves additional factors including the loss myosin ring bisects the plane of the elongating anaphase The checkpoint that delays 52,81 anaphase onset until of actin-binding proteins from cell poles and the ­spindle. Given the speed at which animal cells change 82–84 chromosomes are properly recruitment of septins and BAR-domain proteins to their shape (elongation and cleavage furrow formation) attached to the the site of actomyosin ring formation, where they aid during mitotic exit, this requires active crosstalk between metaphase spindle. membrane deformation85,86. the spindle and the cortex (see REF. 87 for information Structural changes at mitotic exit begin with elong­ about the duration of these signals). This contrasts with ation of the spherical mitotic cells, which now adopt a the situation during mitotic rounding, when changes more elliptical shape. This is followed by cleavage furrow in cell shape and actin organization seem to be regu­ lated relatively independently of microtubules and microtubule-­based structures13 (an exception being a a Plasma membrane potential role for the cortex in centrosome separation at 9,88,89 Chromosome mitotic entry ). Similarly, spindle assembly seems to Microtubule be largely independent of the presence of actin filaments Centrosome both in vivo9 and in vitro90. This difference implies that mitotic exit must be accompanied by profound changes Retraction fibre in spindle–cortex communication — as has long been appreciated (we suggest reading Rappaport’s book87 for an in-depth review of the early literature). Insights into the mechanism came from Rappaport, who carried­ out a series of seminal experiments showing how over­ lapping antiparallel astral microtubules can direct the local formation of an ectopic furrow during mitotic Substrate Actin exit87. Although ectopic furrows do not form in analo­ Myosin 91 pERM gous experiments in all systems , in most animal cells tested, the overlapping microtubules that form between b the poles of the anaphase spindle92 direct the formation and position of the actomyosin furrow to ensure that the division plane bisects the spindle. At a molecular level, this is achieved through recruit­ Adherens centralspindlin complex junction ment of the , a set of proteins that orchestrate the ensuing molecular events92. In brief, overlapping antiparallel microtubules of the anaphase spindle recruit, and are further organized by, a set of kinesin motors (reviewed in detail in REF. 76). Following the APC-induced fall in CDK1–cyclin B activity, these in turn recruit a scaffolding protein, RacGAP1 (also known as MgcRacGAP) together with the RhoGEF ECT2 to the spindle midzone. This shift in ECT2 localization leads to the repolarization of the cortical actomyosin network (and associated proteins such as Anillin93), through the local activation of RhoA94,95. RacGAP1 facilitates ECT2 in the activation of RhoA96, possibly by inactivating Figure 3 | Forces experienced by the cells duringNature mitotic Reviews rounding. | Molecular An Cellisolated Biology cell RacGTP at the midzone97, and therefore helps to estab­ dividing in culture (a) and a cell dividing in an (b) experience a plethora of lish a gradient of Rac and ARP2/3 activity — low at forces (indicated by arrows). At metaphase, these are balanced to give cells their the midzone and high at the poles — aiding anaphase characteristic stable rounded shape. Cells retain retraction fibres, which tether them in elong­ation. In some cells, the repolarization of cortical place. Osmotic swelling causes an increase in the internal pressure (purple arrows), actomyosin may be further augmented by the establish­ which is counteracted by a contractile actomyosin network (red arrows). The actomyosin 98 network is tethered to the cortex by active (phosphorylated) ERM (Ezrin, Radixin, Moesin) ment of a PIP2/PIP3 gradient in anaphase (PIP2 high at proteins (pERM; yellow). Cortically localized dynein motors then pull on astral the furrow, facilitating Rho activation; and PIP3 high microtubules to align the spindle and to determine the plane of cell division. In at the poles, facilitating Rac activation). Finally, this pro­ comparison to cells in culture, cells in an epithelium (b) have additional complexities, as cess is promoted by the local action of the mitotic kinases they are surrounded by neighbouring cells that constantly push back on the dividing cell PLK1 and Aurora B94,99 (a component of the chromosomal (black arrows). In addition, they retain their adherens junctions during cell division. passenger complex (CPC)), which persist at the midzone.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 5 ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

Anaphase-promoting Until recently, it was thought that this same set of Polar relaxation. Physical models of cytokinesis suggest complex cues emanating from the spindle positions the furrow in that the essential factor required for a successful divi­ (APC). An E3 ubiquitin ligase the same way across all animal cell divisions. However, sion is the establishment of a gradient of cortical con­ that targets specific cell cycle spindle–cortex crosstalk is flexible. Thus, asymmetries tractility across the cell76, whereby the cortex is more proteins for degradation by 100–102 the 26S proteasome upon in the cortex can bias the site of furrow formation . contractile at the centre and relatively compliant at cell satisfaction of the spindle In addition, pre-existing cortical asymmetries can mod­ poles. Thus, the assembly and constriction of a central assembly checkpoint, to trigger ify the rate of anaphase elongation101 to alter the size of actomyosin ring may not be sufficient to drive cyto­ the separation of sister daughter cells. Although the instances identified so far kinesis in cells with a rigid mitotic cortex52,104. In line chromatids and the transition are not as extreme as that in budding yeast, in which the with this, division involves both the assembly of an to anaphase. spindle is moved so that it comes to lie across the bud actomyosin ring, induced by overlapping microtubules neck103, it is clear from these few instances that the actin- that are positioned between spindle poles (FIG. 4b,c), based cortex can contribute to the positioning of the and concomitant softening of the cortical actomyosin division plane. network at cell poles105,106 — a process we term polar

a Centralspindlin complex b CPC Actin Plasma membrane pMyosin pERM Sister chromatid Anillin Kinetochore Microtubule Centrosome

PP1–SDS22

Retraction fibre

Substrate PP1–SDS22 PP1–SDS22

Aurora B Aurora B c d Actin

Centralspindlin complex CPC Centrosome PP1 ERM Microtubule (inactive) Sister chromatid

Myosin Microtubule

Plasma membrane Plasma membrane Anillin pERM RhoA Actin

Figure 4 | Polar relaxation and coupling of chromosome segregation to with PP1 dominating at the poles Natureand the Reviews CPC dominating | Molecular at theCell midzone. Biology cytokinesis. a | As sister chromatids separate and cyclin-dependent kinase 1 This, together with the local RanGTP gradient generated by the association (CDK1)–cyclin B activity declines at the onset of anaphase, the of the Ran GEF RCC1 (regulator of chromosome condensation 1) with centralspindlin complex and the chromosomal passenger complex (CPC; mitotic chromatin, probably facilitates the local dephosphorylation of a complex containing active Aurora B) move to the spindle midzone. non-muscle myosin II and ERM (Ezrin, Radixin, Moesin) proteins and induces Centralspindlin recruits ECT2 to the spindle midzone. By contrast, the the loss of Anillin at cell poles, establishing a gradient of actomyosin phosphatase PP1 (in complex with SDS22), remains on the kinetochores. As a across the cell that helps to drive cell division. c | High magnification view of result, the activity domain of the phosphatase and kinase begin to separate the spindle midzone showing the localization of CPC and centralspindilin in space. b | As the sister chromatids begin to approach the poles, the cell complex to the overlapping microtubules, where they induce the local elongates. Anaphase cell elongation is regulated by a combination of forces formation of a contractile actomyosin ring. d | High magnification view of the and signals, many of which emanate from the anaphase spindle. Following cell poles during anaphase. Kinetochore-localized PP1–SDS22 chromosome separation, the activity gradients of the CPC and PP1 separate approaches cell poles at mid-anaphase leading to local dephosphorylation to oppose one another, leading to an opposing kinase–phosphatase gradient of active ERM proteins at the cortex, polar relaxation and local blebbing.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrm ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

relaxation (FIG. 4b,d). Depending on the relative timing In support of this idea, the approach of kinetochores of this local polar loss of phosphorylated ERM (pERM) carrying PP1 and SDS22 (REF. 106) to cell poles is accom­ proteins, actin and non-muscle myosin II, cell elong­ panied by PP1–SDS22‑dependent dephosphorylation ation can be a smooth and gradual process, or it can be of ERM proteins51,52,81 and the loss of polar actomyo­ accompanied by furious rounds of blebbing — as physi­ sin118, before the accumulation of actomyosin at the cell cal connections between the plasma membrane and the midzone. As PP1 promotes the large-scale reversal of underlying cortical actin network are repeatedly lost mitotic phosphorylation at mitotic exit119, potentially and re-­established105. Moreover, failure to induce timely aiding inactivation of the spindle checkpoint120, the rise relaxation of the polar cortex can lead to catastrophic in PP1 activity at anaphase effectively couples cortical failure of cell division105. mechanics (that lead to the shape changes) to the change Three signals have been identified that may contrib­ in mitotic state. In addition, as the kinases that oppose ute to relaxation of the cortex at opposing cell poles PP1 activity (notably Aurora B and Polo kinase) remain (FIG. 4). First, polar relaxation is likely to occur as an at the centre of the spindle during anaphase, where they ­indirect consequence of the movement of the acto­ localize to regions of microtubule overlap (reviewed in myosin network from cell poles towards the central REFS 121,122), the anaphase spindle may set up a phos­ region of the furrow at anaphase105. Anaphase cell phorylation gradient across the cell — with high levels elong­ation has been shown to depend on ROK and non-­ of kinase-mediated phosphorylation near the spindle muscle myosin II activity in fly cells69,107, both of which midzone and high levels of PP1–SDS22 mediated are also required for furrow formation, indicating an dephosphorylation towards the cell poles. In principle, association between the two processes. In addition, polar such a phosphorylation gradient could bridge the physi­ relaxation may be induced by signals that emanate from cal gaps between the spindle, the cytoplasm and the cor­ the anaphase spindle and are independent of overlapping tex, thereby helping to coordinate events across the cell microtubules and centralspindlin. In previous decades, and promoting polarization of the cortical actomyosin this type of activity was often attributed to the ability of before cytokinesis. the growing, plus ends of astral microtubules to induce cortical softening at cell poles108. Although the complex­ Completion of division ity of microtubule-based signals are yet to be resolved At this stage, the contraction of the centrally positioned (for example, furrow components accumulate at the tips actomyosin ring transforms the cell into a dumbbell Actomyosin contractile ring of microtubules in the absence of micro­tubule overlap shape. Although this ring usually closes in a symmetrical A contractile structure in cells exiting mitosis with a monopolar spindle­ 109,110), fashion in cells in culture (FIG. 5a), the ring moves apically composed of actin and myosin it is now clear that the anaphase spindle106,111–113 also as it constricts in many epithelia83,84,123 (FIG. 5b). In some filaments that deforms the plasma membrane to drive signals to the cortex via microtubule-independent sig­ fly epithelia, this basal-to-apical movement of the ring cytokinesis. nals. In support of this, polar relaxation still occurs may be a consequence of the lack of actomyosin at the near anaphase chromatin in cells that lack spindle poles apical surface of mitotic cells14. In addition, it has been Centralspindilin complex and/or microtubules106. suggested that this polarized geometry is a simple con­ A protein complex that Recent work suggests that one or more of the signals sequence of tissue mechanics, as the contractile forces localizes to overlapping antiparallel microtubules in may be chromatin-based. In one study in human cells in that neighbouring cells exert on mitotic cells through anaphase to recruit proteins culture, this was attributed to the action of RCC1 (regu­ apically positioned cell–cell junctions resist ring closure that drive local actomyosin ring lator of chromosome condensation 1), a Ran GEF112. in fly epithelia83,84,124. The opposing forces of ring clo­ formation, so that closure of It was postulated that the association of RCC1 with sure and tissue tension have other consequences. In fly the ring bisects the spindle. mitotic chromatin establishes a local RanGTP gradient epithelia, they physically uncouple the actomyosin ring Chromosomal passenger in anaphase that is high at cell poles and low at the cell and the associated plasma membrane from the existing complex centre. RanGTP then functions via importin‑β to influ­ junction (FIG. 5b), leading, eventually, to the formation (CPC). A protein complex ence the activity and/or localization of a wide range of of a straight, new planar interface between the two consisting of kinase Aurora B actin regulators that carry an NLS (nuclear localization ­daughter cells123. The formation of this new interface and its regulatory subunits 111,114 115 borealin, INCENP and survivin, signal) . These include Anillin , which is lost from may be aided by the reactivation of the ARP2/3 com­ 123 which relocalize from the polar cortex following the approach of chromo­ plex at the cortex close to the midbody , through the kinetochores to overlapping somes112, leading to weakening of the mitotic cortical delivery of membrane from vesicular stores125,126 and/or antiparallel microtubules at the actomyosin network (as is the case during meiosis)116. through the generation of pushing forces on opposing midzones of cells in anaphase, 123 to regulate actomyosin ring This RanGTP pathway, it has been argued, aids spindle membranes . At the same time, these forces may help formation and, later, positioning by negatively regulating local recruitment of trigger abscission, as a loss of tension across the division abscission. nuclear mitotic apparatus protein (NuMA), LGN111 and site has been shown to be a prerequisite for the physical Anilllin112 when anaphase chromatin in an off-centre separation of animal cells in culture127. Midbody spindle approaches one cell pole more closely than the The completion of the division process also Also called a Flemming body, the midbody is a structure that other, to ensure equal partitioning of cell mass during requires disassembly of the actomyosin ring and the connects the two daughter symmetric divisions. microtubule-­rich midbody. This is associated with cells towards the end of In a separate study it was proposed that a kinetochore-­ the binding of RacGAP1 to the membrane, the recruit­ cytokinesis. It controls localized phosphatase117, PP1, functions together with a ment of FIP3 (also known as NEMO) in place of abscission, a process that (REF. 128) 129 leads to the physical regulatory subunit, SDS22, to promote the loss of cor­ ECT2 , ECT2 degradation , and the loss of (REF. 130) 98,131 separation of the two tical actomyosin network from the poles of cells dur­ PIP2 , Rho and actin from the midzone . daughter cells. ing mid-anaphase, helping to trigger polar relaxation. Following disassembly of the microtubule-based

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 7 ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

a Actin Anillin Myosin RhoA pERM ESCRTIII Plasma membrane Microtubule Centrosome Chromatin

Abscission

Substrate

Daughter cells Chromatin Centrosome Microtubule Plasma membrane

Substrate Adhesion

Cell junction b Centrosome Microtubule Chromatin

↑ ARP2/3

Figure 5 | Structural changes at mitotic exit. a | During mitotic exit, the actomyosin network used to drive mitotic Nature Reviews | Molecular Cell Biology rounding is remodelled to form an actomyosin ring that is used to drive cytokinesis. As cells exit from mitosis, the nuclear lamina begins to reform from the poles, thus allowing time for lagging chromosomes to become incorporated into the nascent nucleus. ESCRTIII-mediated abscission leads to the formation of two daughter cells, which form new cell– substrate adhesions and re-spread following abscission. b | In cells dividing in an epithelium, the actomyosin ring moves from basal to apical as it contracts. Additionally, these cells need to form a new cell–cell interface. This may be aided by reactivation of ARP2/3 complex, which pushes adjacent cell membranes together. Finally abscission mediated by ESCRTIII leads to the formation of two daughter cells.

midbody, which is regulated by centrosomes132, mitotic changes in the cell. Further, proteins containing an NLS kinases and Spastin133,134, ESCRTIII is recruited to the — such as Anillin, RacGAP1 and ECT2 — that drive thin membrane tube that connects daughter cells, actin-dependent changes in mitotic cell shape can then where it triggers abscission as cells enter G1 (reviewed return to the nucleus to re‑establish interphase cell in REF. 134). An abscission checkpoint has been pro­ organization. Simultaneously, an increase in the activ­ posed to function as an additional layer of control to ity of RAP1 is thought to promote the assembly of cell–­ coordinate division with chromosome segregation, as substrate adhesions12. This involves Aurora B‑mediated lagging chromosomes act through Aurora B to delay phosphorylation of targets including FHOD1 ESCRTIII-mediated abscission135,136. (FH1/FH2 domain-containing protein 1; a formin) and Finally, as cells return to an interphase state, the the microtubule-binding protein EB3 (also known as nuclear–cytoplasmic compartment boundary is re‑­ MAPRE3)139,140, which promote cell re‑spreading. established through the reassembly of nuclear pores and ESCRTIII-mediated remodelling of the nuclear Conclusions and perspectives envelope137. In addition to its other effects138, this Cell division necessitates a rapid, near-complete restoration­ of the nuclear compartment boundary is remodel­ling of cell shape, structure and polarity, which likely to insulate the cortex from the direct influence of partitions the entire suite of cellular components into chromatin-­based signals to prevent further structural two daughter cells. This is an incredibly complex

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrm ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

physical process, especially for animal cells dividing Although some of the events of cell division are now in the context of a multicellular tissue, and it must be relatively well understood, much remains to be discov­ precisely regulated. During entry into mitosis, events ered. The precise mechanisms underlying spindle–cortex such as adhesion remodelling, assembly of a cortical crosstalk remains an interesting and unsolved problem. actomyosin network, microtubule remodelling and In addition, little is known about the mechanisms that are spindle morphogenesis are coordinated by entrain­ used to ensure that mitosis and cell division are mechani­ ment to the same clock — that is, increasing levels of cally robust62 and that the various organelles are precisely mitotic kinase activity — as well as by the dramatic segregated during division142–144, or about the mech­ biochemical changes that accompany the sudden loss anisms that are used to bias these processes to achieve of the compartment boundary that separates the cyto­ the same precision during asymmetric divisions. Finally, plasm and nucleus at the transition from prophase to very little is known about the evolution of eukary­otic cell prometaphase. Thus, for most purposes, the micro­ division, which appears to have its origins in Archaea2,145. tubule and actin-based structures that are essential for As progression through mitosis is accompanied by rapid, chromosome segregation and cell division, respectively, sequential and wholesale changes in cell structure, the are constructed in parallel. Nevertheless, a successful study of cell division provides researchers with an ideal division requires spatial coordination of the cortex and window into the dynamic regulation of cell architecture. the spindle, necessitating crosstalk between these two The bulk of these events are driven by changes in the systems. This communication is essential to ensure that activities of a small set of kinases and phosphatases, the metaphase spindle is properly oriented with respect ­making it relatively easy to identify the switches and to interphase and mitotic cell shape, cell–cell junctions molecules that underpin these changes. Moreover, this and tissue tension; and that the cytokinesis ring is posi­ type of research is being aided by the advent of live tioned so that it bisects the anaphase spindle. Finally, super-­resolution microscopy, which allows changes the return to interphase organization also requires in cell architecture to be imaged at an unprecedented­ both temporal (cyclin degradation, the loss of mitotic level, together with CRISPR–Cas9‑mediated genome kinase activity and the increasing dominance of mitotic editing and opto­genetics, which provide new tools to phosphatases) and spatial (gradients of activity and re‑­ test hypotheses and reveal the underlying molecu­lar establishment of compartment boundaries) control. As mechanisms. Therefore, although the mass of cell divi­ an example of this, the nuclear envelope seals from the sion literature (only some of which we have been able to poles to the centre138, enabling lagging chromosomes summarize here) can seem overwhelming and the field to be gathered up into daughter nuclei to prevent the saturated, there are rich untapped seams waiting to be formation of micronuclei and chromothripsis141. mined and the perfect tools with which to do it.

1. Jongsma, M. L., Berlin, I. & Neefjes, J. On the move: oriented along the long cell axis. Proc. Natl Acad. 21. Nigg, E. A. The substrates of the cdc2 kinase. organelle dynamics during mitosis. Trends Cell Biol. Sci. USA 112, 5726–5731 (2015). Semin. Cell Biol. 2, 261–270 (1991). 25, 112–124 (2015). 12. Dao, V. T., Dupuy, A. G., Gavet, O., Caron, E. 22. Curtis, M., Nikolopoulos, S. N. & Turner, C. E. 2. Balasubramanian, M. K., Srinivasan, R., Huang, Y. & de Gunzburg, J. Dynamic changes in Rap1 Actopaxin is phosphorylated during mitosis and is a & Ng, K. H. Comparing contractile apparatus-driven activity are required for cell retraction and substrate for cyclin B1/cdc2 kinase. Biochem. J. 363, cytokinesis mechanisms across kingdoms. spreading during mitosis. J. Cell Sci. 122, 233–242 (2002). Cytoskeleton 69, 942–956 (2012). 2996–3004 (2009). 23. Olsen, J. V. et al. Quantitative phosphoproteomics 3. Mercier, R., Kawai, Y. & Errington, J. General This study identifies RAP1 as a key regulator reveals widespread full phosphorylation site principles for the formation and proliferation of a of mitotic adhesion remodelling. occupancy during mitosis. Sci. Signal. 3, ra3 (2010). wall‑free (L‑form) state in bacteria. eLife 3, e04629 13. Matthews, H. K. et al. Changes in Ect2 localization 24. Suzuki, K. & Takahashi, K. Reduced cell adhesion (2014). couple actomyosin-dependent cell shape changes during mitosis by threonine phosphorylation of 4. Nestor-Bergmann, A., Goddard, G. & Woolner, S. to mitotic progression. Dev. Cell 23, 371–383 β1 integrin. J. Cell. Physiol. 197, 297–305 (2003). Force and the spindle: mechanical cues in mitotic (2012). 25. Yamakita, Y. et al. Dissociation of FAK/p130CAS/c‑Src spindle orientation. Semin. Cell Dev. Biol. 34, This study identifies ECT2 as a master regulator of complex during mitosis: role of mitosis-specific serine 133–139 (2014). cell shape changes, during both entry into and exit phosphorylation of FAK. J. Cell Biol. 144, 315–324 5. Fink, J. et al. External forces control mitotic from mitosis. (1999). spindle positioning. Nat. Cell Biol. 13, 771–778 14. Rosa, A., Vlassaks, E., Pichaud, F. & Baum, B. 26. Cramer, L. P. & Mitchison, T. J. Investigation of the (2011). Ect2/Pbl acts via Rho and polarity proteins to mechanism of retraction of the cell margin and 6. Stout, J. R., Rizk, R. S., Kline, S. L. & Walczak, C. E. direct the assembly of an isotropic actomyosin rearward flow of nodules during mitotic cell rounding. Deciphering protein function during mitosis in PtK cortex upon mitotic entry. Dev. Cell 32, 604–616 Mol. Biol. Cell 8, 109–119 (1997). cells using RNAi. BMC Cell Biol. 7, 26 (2006). (2015). This study was one of the few to study the 7. Woolner, S. & Papalopulu, N. Spindle position in 15. Gavet, O. & Pines, J. Progressive activation of formation of retraction fibres during mitotic symmetric cell divisions during epiboly is controlled by CyclinB1‑Cdk1 coordinates entry to mitosis. Dev. Cell rounding. opposing and dynamic apicobasal forces. Dev. Cell 22, 18, 533–543 (2010). 27. Toyoshima, F. & Nishida, E. Integrin-mediated 775–787 (2012). 16. Solomon, M. J., Glotzer, M., Lee, T. H., Philippe, M. adhesion orients the spindle parallel to the 8. Zang, J. H. et al. On the role of myosin‑II in & Kirschner, M. W. Cyclin activation of p34cdc2. Cell substratum in an EB1- and myosin X‑dependent cytokinesis: division of Dictyostelium cells under 63, 1013–1024 (1990). manner. EMBO J. 26, 1487–1498 (2007). adhesive and nonadhesive conditions. Mol. Biol. Cell 17. Pomerening, J. R., Sontag, E. D. & Ferrell, J. E. Jr. 28. Mali, P., Wirtz, D. & Searson, P. C. Interplay of RhoA 8, 2617–2629 (1997). Building a cell cycle oscillator: hysteresis and and motility in the programmed spreading of 9. Lancaster, O. M. et al. Mitotic rounding alters cell bistability in the activation of Cdc2. Nat. Cell Biol. 5, daughter cells postmitosis. Biophys. J. 99, geometry to ensure efficient bipolar spindle formation. 346–351 (2003). 3526–3534 (2010). Dev. Cell 25, 270–283 (2013). 18. Pomerening, J. R., Kim, S. Y. & Ferrell, J. E. Jr. 29. Ferreira, J. G., Pereira, A. J., Akhmanova, A. This study shows that mitotic rounding (rather than Systems-level dissection of the cell-cycle oscillator: & Maiato, H. Aurora B spatially regulates EB3 the actin cytoskeleton) is important for spindle bypassing positive feedback produces damped phosphorylation to coordinate daughter cell adhesion formation. oscillations. Cell 122, 565–578 (2005). with cytokinesis. J. Cell Biol. 201, 709–724 (2013). 10. Strzyz, P. J. et al. Interkinetic nuclear migration is 19. Robertson, J. et al. Defining the phospho-adhesome 30. Bellis, J. et al. The tumor suppressor Apc controls centrosome independent and ensures apical cell through the phosphoproteomic analysis of integrin planar cell polarities central to gut homeostasis. division to maintain tissue integrity. Dev. Cell 32, signalling. Nat. Commun. 6, 6265 (2015). J. Cell Biol. 198, 331–341 (2012). 203–219 (2015). 20. Marchesi, S. et al. DEPDC1B coordinates de‑adhesion 31. Kosodo, Y. et al. Cytokinesis of neuroepithelial cells 11. Wyatt, T. P. et al. Emergence of homeostatic epithelial events and cell-cycle progression at mitosis. Dev. Cell can divide their basal process before anaphase. packing and stress dissipation through divisions 31, 420–433 (2014). EMBO J. 27, 3151–3163 (2008).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 9 ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

32. Reinsch, S. & Karsenti, E. Orientation of spindle axis 56. Maniti, O. et al. Binding of moesin and ezrin to cleavage furrow formation during cytokinesis. Dev. Cell and distribution of plasma membrane proteins during membranes containing phosphatidylinositol (4,5) 21, 1104–1115 (2011). cell division in polarized MDCKII cells. J. Cell Biol. bisphosphate: a comparative study of the affinity This study shows how ECT2 is localized to the 126, 1509–1526 (1994). constants and conformational changes. plasma membrane at the cell midzone at the 33. Lecuit, T. & Wieschaus, E. Junctions as organizing Biochim. Biophys. Acta 1818, 2839–2849 (2012). onset of anaphase. centers in epithelial cells? A fly perspective. Traffic 3, 57. Zhai, Y., Kronebusch, P. J., Simon, P. M. & Borisy, G. G. 81. Kunda, P. et al. PP1‑mediated moesin 92–97 (2002). Microtubule dynamics at the G2/M transition: abrupt dephosphorylation couples polar relaxation to mitotic 34. Clark, A. G., Dierkes, K. & Paluch, E. K. Monitoring breakdown of cytoplasmic microtubules at nuclear exit. Curr. Biol. 22, 231–236 (2012). actin cortex thickness in live cells. Biophys. J. 105, envelope breakdown and implications for spindle 82. Mavrakis, M. et al. Septins promote F‑actin ring 570–580 (2013). morphogenesis. J. Cell Biol. 135, 201–214 (1996). formation by crosslinking actin filaments into curved 35. Bovellan, M. et al. Cellular control of cortical actin 58. Clarke, P. R. & Zhang, C. Spatial and temporal bundles. Nat. Cell Biol. 16, 322–334 (2014). nucleation. Curr. Biol. 24, 1628–1635 (2014). coordination of mitosis by Ran GTPase. Nat. Rev. Mol. 83. Founounou, N., Loyer, N. & Le Borgne, R. Septins This study identifies roles for different actin Cell Biol. 9, 464–477 (2008). regulate the contractility of the actomyosin ring to regulators in cortical actin network formation. 59. Stewart, M. P. et al. Hydrostatic pressure and the enable adherens junction remodeling during 36. Castanon, I. et al. Anthrax toxin receptor 2a controls actomyosin cortex drive mitotic cell rounding. Nature cytokinesis of epithelial cells. Dev. Cell 24, 242–255 mitotic spindle positioning. Nat. Cell Biol. 15, 28–39 469, 226–230 (2011). (2013). (2013). This study was the first to propose a role for 84. Guillot, C. & Lecuit, T. Adhesion disengagement 37. Ibarra, N., Pollitt, A. & Insall, R. H. Regulation of actin changes in osmotic pressure in generating the uncouples intrinsic and extrinsic forces to drive assembly by SCAR/WAVE proteins. Biochem. Soc. forces accompanying mitotic rounding. cytokinesis in epithelial tissues. Dev. Cell 24, Trans. 33, 1243–1246 (2005). 60. Son, S. et al. Resonant microchannel volume and mass 227–241 (2013). 38. Davidson, A. J., Ura, S., Thomason, P. A., Kalna, G. measurements show that suspended cells swell during 85. Takeda, T. et al. Drosophila F‑BAR protein Syndapin & Insall, R. H. Abi is required for modulation and mitosis. J. Cell Biol. 211, 757–763 (2015). contributes to coupling the plasma membrane and stability but not localization or activation of the 61. Zlotek-Zlotkiewicz, E., Monnier, S., Cappello, G., contractile ring in cytokinesis. Open Biol. 3, 130081 SCAR/WAVE complex. Eukaryot. Cell 12, 1509–1516 Le Berre, M. & Piel, M. Optical volume and mass (2013). (2013). measurements show that mammalian cells swell 86. Roberts-Galbraith, R. H. et al. Dephosphorylation of 39. Zhuang, C. et al. CDK1‑mediated phosphorylation during mitosis. J. Cell Biol. 211, 765–774 (2015). F‑BAR protein Cdc15 modulates its conformation of Abi1 attenuates Bcr-Abl-induced F‑actin assembly This study shows that cells swell as they enter and stimulates its scaffolding activity at the cell and tyrosine phosphorylation of WAVE complex mitosis. division site. Mol. Cell 39, 86–99 (2010). during mitosis. J. Biol. Chem. 286, 38614–38626 62. Cattin, C. J. et al. Mechanical control of mitotic 87. Rappaport, R. Cytokinesis in Animal Cells (2011). progression in single animal cells. Proc. Natl Acad. (Cambridge Univ. Press, 1996). 40. Skau, C. T., Neidt, E. M. & Kovar, D. R. Role of Sci. USA 112, 11258–11263 (2015). This book summarizes decades of cell division tropomyosin in formin-mediated contractile ring 63. Sorce, B. et al. Mitotic cells contract actomyosin cortex research. assembly in fission yeast. Mol. Biol. Cell 20, and generate pressure to round against or escape 88. Rosenblatt, J., Cramer, L. P., Baum, B. & McGee, K. M. 2160–2173 (2009). epithelial confinement. Nat. Commun. 6, 8872 (2015). Myosin II‑dependent cortical movement is required for 41. Miki, T., Smith, C. L., Long, J. E., Eva, A. & 64. Ramanathan, S. P. et al. Cdk1‑dependent mitotic centrosome separation and positioning during mitotic Fleming, T. P. Oncogene ect2 is related to regulators enrichment of cortical myosin II promotes cell spindle assembly. Cell 117, 361–372 (2004). of small GTP-binding proteins. Nature 362, 462–465 rounding against confinement. Nat. Cell Biol. 17, 89. De Simone, A., Nedelec, F. & Gonczy, P. Dynein (1993). 148–159 (2015). transmits polarized actomyosin cortical flows to 42. Maddox, A. S. & Burridge, K. RhoA is required for 65. Fischer-Friedrich, E., Hyman, A. A., Julicher, F., promote centrosome separation. Cell Rep. 14, cortical retraction and rigidity during mitotic cell Muller, D. J. & Helenius, J. Quantification of surface 2250–2262 (2016). rounding. J. Cell Biol. 160, 255–265 (2003). tension and internal pressure generated by single 90. Sawin, K. E. & Mitchison, T. J. Mitotic spindle This study identifies RhoA as a key regulator mitotic cells. Sci. Rep. 4, 6213 (2014). assembly by two different pathways in vitro. J. Cell of mitotic rounding. 66. Hoijman, E., Rubbini, D., Colombelli, J. & Alsina, B. Biol. 112, 925–940 (1991). 43. Yuce, O., Piekny, A. & Glotzer, M. An ECT2– Mitotic cell rounding and epithelial thinning regulate 91. Mitchison, T. et al. Growth, interaction, and centralspindlin complex regulates the localization and lumen growth and shape. Nat. Commun. 6, 7355 positioning of microtubule asters in extremely large function of RhoA. J. Cell Biol. 170, 571–582 (2005). (2015). vertebrate embryo cells. Cytoskeleton 69, 738–750 44. Watanabe, N., Kato, T., Fujita, A., Ishizaki, T. 67. Kondo, T. & Hayashi, S. Mitotic cell rounding (2012). & Narumiya, S. Cooperation between mDia1 accelerates epithelial invagination. Nature 494, 92. White, E. A. & Glotzer, M. Centralspindlin: at the heart and ROCK in Rho-induced actin reorganization. 125–129 (2013). of cytokinesis. Cytoskeleton 69, 882–892 (2012). Nat. Cell Biol. 1, 136–143 (1999). This study shows how the forces generated by 93. Piekny, A. J. & Glotzer, M. Anillin is a scaffold protein 45. Oceguera-Yanez, F. et al. Ect2 and MgcRacGAP mitotic rounding can buckle a tissue. that links RhoA, actin, and myosin during cytokinesis. regulate the activation and function of Cdc42 in 68. McCarthy Campbell, E. K., Werts, A. D. & Goldstein, B. Curr. Biol. 18, 30–36 (2008). mitosis. J. Cell Biol. 168, 221–232 (2005). A cell cycle timer for asymmetric spindle positioning. 94. Nguyen, P. A. et al. Spatial organization of cytokinesis 46. Motegi, F. & Sugimoto, A. Sequential functioning of PLoS Biol. 7, e1000088 (2009). signaling reconstituted in a cell-free system. Science the ECT‑2 RhoGEF, RHO‑1 and CDC‑42 establishes cell 69. Echard, A. & O’Farrell, P. H. The degradation of two 346, 244–247 (2014). polarity in Caenorhabditis elegans embryos. Nat. Cell mitotic cyclins contributes to the timing of cytokinesis. 95. Bement, W. M., Benink, H. A. & von Dassow, G. Biol. 8, 978–985 (2006). Curr. Biol. 13, 373–383 (2003). A microtubule-dependent zone of active RhoA during 47. Field, C. M. et al. Actin behavior in bulk cytoplasm is 70. Mossaid, I. & Fahrenkrog, B. Complex commingling: cleavage plane specification. J. Cell Biol. 170, 91–101 cell cycle regulated in early vertebrate embryos. J. Cell nucleoporins and the spindle assembly checkpoint. (2005). Sci. 124, 2086–2095 (2011). Cells 4, 706–725 (2015). 96. Zhang, D. & Glotzer, M. The RhoGAP activity of CYK‑4/ This study shows how cell cycle state influences 71. Doncic, A., Ben-Jacob, E. & Barkai, N. Evaluating MgcRacGAP functions non-canonically by promoting actin dynamics. putative mechanisms of the mitotic spindle RhoA activation during cytokinesis. eLife 4, e08898 48. Ritchey, L. & Chakrabarti, R. Aurora A kinase checkpoint. Proc. Natl Acad. Sci. USA 102, (2015). modulates actin cytoskeleton through phosphorylation 6332–6337 (2005). 97. Bastos, R. N., Penate, X., Bates, M., Hammond, D. of Cofilin: implication in the mitotic process. 72. Foley, E. A. & Kapoor, T. M. Microtubule attachment & Barr, F. A. CYK4 inhibits Rac1‑dependent PAK1 and Biochim. Biophys. Acta 1843, 2719–2729 (2014). and spindle assembly checkpoint signalling at the ARHGEF7 effector pathways during cytokinesis. J. Cell 49. Takahashi, H., Funakoshi, H. & Nakamura, T. kinetochore. Nat. Rev. Mol. Cell Biol. 14, 25–37 Biol. 198, 865–880 (2012). LIM‑kinase as a regulator of actin dynamics in (2013). 98. Logan, M. R. & Mandato, C. A. Regulation of the actin spermatogenesis. Cytogenet. Genome Res. 103, 73. Chang, L. & Barford, D. Insights into the anaphase- cytoskeleton by PIP2 in cytokinesis. Biol. Cell 98, 290–298 (2003). promoting complex: a molecular machine that 377–388 (2006). 50. Kuilman, T. et al. Identification of Cdk targets that regulates mitosis. Curr. Opin. Struct. Biol. 29, 1–9 99. Basant, A. et al. Aurora B kinase promotes cytokinesis control cytokinesis. EMBO J. 34, 81–96 (2015). (2014). by inducing centralspindlin oligomers that associate 51. Carreno, S. et al. Moesin and its activating kinase Slik 74. Nasmyth, K. Cohesin: a catenase with separate entry with the plasma membrane. Dev. Cell 33, 204–215 are required for cortical stability and microtubule and exit gates? Nat. Cell Biol. 13, 1170–1177 (2011). (2015). organization in mitotic cells. J. Cell Biol. 180, 75. Sivakumar, S. & Gorbsky, G. J. Spatiotemporal 100. Pacquelet, A., Uhart, P., Tassan, J. P. & Michaux, G. 739–746 (2008). regulation of the anaphase-promoting complex in PAR‑4 and anillin regulate myosin to coordinate 52. Kunda, P., Pelling, A. E., Liu, T. & Baum, B. Moesin mitosis. Nat. Rev. Mol. Cell Biol. 16, 82–94 (2015). spindle and furrow position during asymmetric controls cortical rigidity, cell rounding, and spindle 76. Fededa, J. P. & Gerlich, D. W. Molecular control of division. J. Cell Biol. 210, 1085–1099 (2015). morphogenesis during mitosis. Curr. Biol. 18, 91–101 animal cell cytokinesis. Nat. Cell Biol. 14, 440–447 101. Cabernard, C., Prehoda, K. E. & Doe, C. Q. A spindle- (2008). (2012). independent cleavage furrow positioning pathway. References 51 and 52 identify roles for active ERM 77. Schellhaus, A. K., De Magistris, P. & Antonin, W. Nature 467, 91–94 (2010). proteins in the assembly of the mitotic cortex. Nuclear reformation at the end of mitosis. J. Mol. Biol. 102. Ou, G., Stuurman, N., D’Ambrosio, M. & Vale, R. D. 53. Li, Q. et al. Self-masking in an intact ERM-merlin 428, 1962–1985 (2015). Polarized myosin produces unequal-size daughters protein: an active role for the central α-helical domain. 78. Field, C. M. & Alberts, B. M. Anillin, a contractile ring during asymmetric cell division. Science 330, J. Mol. Biol. 365, 1446–1459 (2007). protein that cycles from the nucleus to the cell cortex. 677–680 (2010). 54. Roubinet, C. et al. Molecular networks linked by J. Cell Biol. 131, 165–178 (1995). These studies show that the position of the division Moesin drive remodeling of the cell cortex during 79. Green, R. A., Paluch, E. & Oegema, K. Cytokinesis plane is not always a simple consequence of spindle mitosis. J. Cell Biol. 195, 99–112 (2011). in animal cells. Annu. Rev. Cell Dev. Biol. 28, 29–58 position. 55. Janetopoulos, C. & Devreotes, P. Phosphoinositide (2012). 103. Yeh, E. et al. Dynamic positioning of mitotic spindles signaling plays a key role in cytokinesis. J. Cell Biol. 80. Su, K. C., Takaki, T. & Petronczki, M. Targeting of the in yeast: role of microtubule motors and cortical 174, 485–490 (2006). RhoGEF Ect2 to the equatorial membrane controls determinants. Mol. Biol. Cell 11, 3949–3961 (2000).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrm ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.

REVIEWS

104. Charras, G. & Paluch, E. Blebs lead the way: how to 120. Lesage, B., Qian, J. & Bollen, M. Spindle checkpoint 135. Steigemann, P. et al. Aurora B‑mediated abscission migrate without lamellipodia. Nat. Rev. Mol. Cell Biol. silencing: PP1 tips the balance. Curr. Biol. 21, checkpoint protects against tetraploidization. Cell 9, 730–736 (2008). R898–R903 (2011). 136, 473–484 (2009). 105. Sedzinski, J. et al. Polar actomyosin contractility 121. Petronczki, M., Lenart, P. & Peters, J. M. Polo on 136. Carlton, J. G., Caballe, A., Agromayor, M., Kloc, M. destabilizes the position of the cytokinetic furrow. the rise — from mitotic entry to cytokinesis with Plk1. & Martin-Serrano, J. ESCRT-III governs the Nature 476, 462–466 (2011). Dev. Cell 14, 646–659 (2008). Aurora B‑mediated abscission checkpoint through 106. Rodrigues, N. T. et al. Kinetochore-localized 122. Kitagawa, M. & Lee, S. H. The chromosomal CHMP4C. Science 336, 220–225 (2012). PP1–Sds22 couples chromosome segregation to passenger complex (CPC) as a key orchestrator of 137. Olmos, Y., Hodgson, L., Mantell, J., Verkade, P. polar relaxation. Nature 524, 489–492 (2015). orderly mitotic exit and cytokinesis. Front. Cell Dev. & Carlton, J. G. ESCRT-III controls nuclear envelope These studies investigate the regulation and Biol. 3, 14 (2015). reformation. Nature 522, 236–239 (2015). function of anaphase polar relaxation. 123. Herszterg, S., Leibfried, A., Bosveld, F., Martin, C. 138. Maiato, H., Afonso, O. & Matos, I. A chromosome 107. Hickson, G. R., Echard, A. & O’Farrell, P. H. Rho-kinase & Bellaiche, Y. Interplay between the dividing cell and separation checkpoint: a midzone Aurora B gradient controls cell shape changes during cytokinesis. its neighbors regulates adherens junction formation mediates a chromosome separation checkpoint that Curr. Biol. 16, 359–370 (2006). during cytokinesis in epithelial tissue. Dev. Cell 24, regulates the anaphase-telophase transition. 108. Wolpert, L. A relaxed modification of the Rappaport 256–270 (2013). Bioessays 37, 257–266 (2015). model for cytokinesis. J. Theor. Biol. 345, 109 This study asks how cell division is organized to 139. Floyd, S. et al. Spatiotemporal organization of Aurora‑B (2014). generate a new cell–cell interface following division by APC/CCdh1 after mitosis coordinates cell spreading 109. Canman, J. C. et al. Determining the position of the in a tissue. through FHOD1. J. Cell Sci. 126, 2845–2856 (2013). cell division plane. Nature 424, 1074–1078 (2003). 124. Morais-de‑Sa, E. & Sunkel, C. E. Connecting polarized 140. Ferreira, J. G., Pereira, A. J., Akhmanova, A. 110. Hu, C. K., Coughlin, M., Field, C. M. & Mitchison, T. J. cytokinesis to epithelial architecture. Cell Cycle 12, & Maiato, H. Aurora B spatially regulates EB3 Cell polarization during monopolar cytokinesis. J. Cell 3583–3584 (2013). phosphorylation to coordinate daughter cell adhesion Biol. 181, 195–202 (2008). 125. Mitsushima, M. et al. Revolving movement of a with cytokinesis. J. Cell Biol. 201, 709–724 (2013). This study explores how the cytokinesis machinery dynamic cluster of actin filaments during mitosis. 141. Crasta, K. et al. DNA breaks and chromosome responds to forced mitotic exit in the absence of J. Cell Biol. 191, 453–462 (2010). pulverization from errors in mitosis. Nature 482, a bipolar spindle. 126. Neto, H., Collins, L. L. & Gould, G. W. Vesicle 53–58 (2012). 111. Kiyomitsu, T. & Cheeseman, I. M. Chromosome- and trafficking and membrane remodelling in cytokinesis. 142. Kressmann, S., Campos, C., Castanon, I., spindle-pole-derived signals generate an intrinsic code Biochem. J. 437, 13–24 (2011). Furthauer, M. & Gonzalez-Gaitan, M. Directional for spindle position and orientation. Nat. Cell Biol. 14, 127. Lafaurie-Janvore, J. et al. ESCRT-III assembly and Notch trafficking in Sara endosomes during 311–317 (2012). cytokinetic abscission are induced by tension release asymmetric cell division in the spinal cord. Nat. Cell 112. Kiyomitsu, T. & Cheeseman, I. M. Cortical dynein in the intercellular bridge. Science 339, 1625–1629 Biol. 17, 333–339 (2015). and asymmetric membrane elongation coordinately (2013). 143. Furthauer, M. & Gonzalez-Gaitan, M. Endocytosis position the spindle in anaphase. Cell 154, 391–402 128. Simon, G. C. et al. Sequential Cyk‑4 binding to ECT2 and mitosis: a two-way relationship. Cell Cycle 8, (2013). and FIP3 regulates cleavage furrow ingression 3311–3318 (2009). 113. Connell, M., Cabernard, C., Ricketson, D., Doe, C. Q. and abscission during cytokinesis. EMBO J. 27, 144. Jajoo, R. et al. Accurate concentration control of & Prehoda, K. E. Asymmetric cortical extension shifts 1791–1803 (2008). mitochondria and nucleoids. Science 351, 169–172 cleavage furrow position in Drosophila neuroblasts. 129. Chalamalasetty, R. B., Hummer, S., Nigg, E. A. (2016). Mol. Biol. Cell 22, 4220–4226 (2011). & Sillje, H. H. Influence of human Ect2 145. Bernander, R. Archaea and the cell cycle. 114. Bird, S. L., Heald, R. & Weis, K. RanGTP and CLASP1 depletion and overexpression on cleavage furrow Mol. Microbiol. 29, 955–961 (1998). cooperate to position the mitotic spindle. Mol. Biol. formation and abscission. J. Cell Sci. 119, 146. Mauser, J. F. & Prehoda, K. E. Inscuteable regulates Cell 24, 2506–2514 (2013). 3008–3019 (2006). the Pins-Mud spindle orientation pathway. PLoS ONE 115. Silverman-Gavrila, R. V., Hales, K. G. & Wilde, A. 130. Dambournet, D. et al. Rab35 GTPase and OCRL 7, e29611 (2012). Anillin-mediated targeting of peanut to phosphatase remodel lipids and F‑actin for 147. Siller, K. H. & Doe, C. Q. Lis1/dynactin regulates pseudocleavage furrows is regulated by the GTPase successful cytokinesis. Nat. Cell Biol. 13, 981–988 metaphase spindle orientation in Drosophila Ran. Mol. Biol. Cell 19, 3735–3744 (2008). (2011). neuroblasts. Dev. Biol. 319, 1–9 (2008). 116. Dehapiot, B. & Halet, G. Ran GTPase promotes oocyte 131. Manukyan, A., Ludwig, K., Sanchez-Manchinelly, S., 148. Zheng, Z. et al. Evidence for dynein and astral polarization by regulating ERM (Ezrin/Radixin/ Parsons, S. J. & Stukenberg, P. T. A complex of microtubule-mediated cortical release and transport

Moesin) inactivation. Cell Cycle 12, 1672–1678 p190RhoGAP‑A and anillin modulates RhoA-GTP of Gαi/LGN/NuMA complex in mitotic cells. Mol. Biol. (2013). and the cytokinetic furrow in human cells. J. Cell Sci. Cell 24, 901–913 (2013). 117. Ohkura, H. & Yanagida, M. S. pombe gene sds22+ 128, 50–60 (2015). essential for a midmitotic transition encodes a leucine- 132. Piel, M., Nordberg, J., Euteneuer, U. & Bornens, M. Acknowledgements rich repeat protein that positively modulates protein Centrosome-dependent exit of cytokinesis in animal The authors would like to thank members of the Baum phosphatase‑1. Cell 64, 149–157 (1991). cells. Science 291, 1550–1553 (2001). laboratory and anonymous reviewers for their critical reading 118. Grusche, F. A. et al. Sds22, a PP1 phosphatase 133. Connell, J. W., Lindon, C., Luzio, J. P. & Reid, E. of the text. B.B. and N.R. thank Cancer Research UK for regulatory subunit, regulates epithelial cell polarity Spastin couples microtubule severing to membrane funding, and B.B. thanks the UK Biotechnology and Biological and shape [Sds22 in epithelial morphology]. BMC Dev. traffic in completion of cytokinesis and secretion. Sciences Research Council (BBSRC) and University College Biol. 9, 14 (2009). Traffic 10, 42–56 (2009). London for support. 119. Wurzenberger, C. & Gerlich, D. W. Phosphatases: 134. Mierzwa, B. & Gerlich, D. W. Cytokinetic abscission: providing safe passage through mitotic exit. Nat. Rev. molecular mechanisms and temporal control. Dev. Cell Competing interests statement Mol. Cell Biol. 12, 469–482 (2011). 31, 525–538 (2014). The authors declare no competing interests.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 11 ©2016 Mac millan Publishers Li mited. All ri ghts reserved. ©2016 Mac millan Publishers Li mited. All ri ghts reserved.