fip3-endosome-dependent formation of the secondary ingression mediates escrt-iii recruitment during...

21
ARTICLES FIP3-endosome-dependent formation of the secondary ingression mediates ESCRT-III recruitment during cytokinesis John A. Schiel 1 , Glenn C. Simon 1 , Chelsey Zaharris 2 , Julie Weisz 3 , David Castle 4 , Christine C. Wu 3 and Rytis Prekeris 1,5 The final cytokinesis event involves severing of the connecting intercellular bridge (ICB) between daughter cells. FIP3-positive recycling endosomes (FIP3 endosomes) and ESCRT complexes have been implicated in mediating the final stages of cytokinesis. Here we analyse the spatiotemporal dynamics of the actin cytoskeleton, FIP3-endosome fusion and ESCRT-III localization during cytokinesis to show that the ICB narrows by a FIP3-endosome-mediated secondary ingression, whereas the ESCRT-III complex is needed only for the last scission step of cytokinesis. We characterize the role of FIP3 endosomes during cytokinesis to demonstrate that FIP3 endosomes deliver SCAMP2/3 and p50RhoGAP to the ICB during late telophase, proteins required for the formation of the secondary ingression. We also show that the FIP3-endosome-induced secondary ingression is required for the recruitment of the ESCRT-III complex to the abscission site. Finally, we characterize a FIP3-endosome-dependent regulation of the ICB cortical actin network through the delivery of p50RhoGAP. These results provide a framework for the coordinated efforts of actin, FIP3 endosomes and the ESCRTs to regulate cytokinesis and abscission. Cytokinesis begins with a primary ingression mediated by a contraction of an actomyosin ring 1 , leading to the formation of the ICB, which is further resolved by thinning of the ICB from 2 μm to 100 nm (secondary ingression) 2 , followed by the ICB plasma membrane fusion (abscission) 2–4 . Recent studies have shown that secondary ingression and abscission involve depolymerization of ICB actin and microtubules 2–4 , fusion of FIP3 endosomes 2,5–7 and recruitment of the ESCRT complexes 8–10 . However, although it was shown that midbody accumulation of ALIX and TSG101 (ESCRT-I) recruits CHMP4B (ESCRT-III) during late cytokinesis 3,8,10 , the roles of FIP3 endosomes and ESCRT complexes during the formation of the secondary ingression and abscission remain to be fully understood. Here, we demonstrate that secondary ingression is mediated by FIP3-endosome fusion and occurs before ESCRT-III recruitment to the abscission site, and that ESCRT-III is required only for stabilization of the formed secondary ingression and final membrane scission. Furthermore, we use time-lapse microscopy and organelle proteomics to identify secretory carrier membrane protein 2/3 (SCAMP2/3) and p50RhoGAP as membrane-traffic- and actin-regulating proteins 1 Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA. 2 Department of Structural Biology and Biophysics, School of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA. 3 Department of Cell Biology and Physiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA. 4 Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA. 5 Correspondence should be addressed to R.P. (e-mail: [email protected]) Received 30 May 2012; accepted 9 August 2012; published online 23 September 2012; DOI: 10.1038/ncb2577 required for abscission, which are delivered to the ICB by FIP3 endosomes. We support these findings by demonstrating that FIP3- endosome delivery of SCAMP2/3 or p50RhoGAP regulates actin depolymerization within the ICB and recruitment of ESCRT-III to the abscission site. RESULTS ESCRT-III recruitment to the midbody coincides with the accumulation of FIP3 endosomes Many reports have shown that FIP3- and Rab11-positive endosomes accumulate at the ICB and are required for abscission 2,6,7,11,12 , and that the movement of FIP3 from centrosomes to the ICB marks the progression of cells from early to late telophase (Supplementary Fig. S1A). A recent study suggested that endosomes are not required for abscission, on the basis of an observation that RAB8 endosomes are not present at the ICB during late cytokinesis 4 . As it is unclear whether RAB8 actually marks FIP3 endosomes, we compared RAB8 and FIP3 during cytokinesis and show that there is little co-localization between RAB8 and FIP3 during early telophase, as RAB8 endosomes are 1068 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 © 2012 Macmillan Publishers Limited. All rights reserved.

Upload: rytis

Post on 21-Feb-2017

217 views

Category:

Documents


3 download

TRANSCRIPT

ART I C L E S

FIP3-endosome-dependent formation of the secondaryingression mediates ESCRT-III recruitmentduring cytokinesisJohn A. Schiel1, Glenn C. Simon1, Chelsey Zaharris2, Julie Weisz3, David Castle4, Christine C. Wu3

and Rytis Prekeris1,5

The final cytokinesis event involves severing of the connecting intercellular bridge (ICB) between daughter cells. FIP3-positiverecycling endosomes (FIP3 endosomes) and ESCRT complexes have been implicated in mediating the final stages of cytokinesis.Here we analyse the spatiotemporal dynamics of the actin cytoskeleton, FIP3-endosome fusion and ESCRT-III localization duringcytokinesis to show that the ICB narrows by a FIP3-endosome-mediated secondary ingression, whereas the ESCRT-III complex isneeded only for the last scission step of cytokinesis. We characterize the role of FIP3 endosomes during cytokinesis todemonstrate that FIP3 endosomes deliver SCAMP2/3 and p50RhoGAP to the ICB during late telophase, proteins required for theformation of the secondary ingression. We also show that the FIP3-endosome-induced secondary ingression is required for therecruitment of the ESCRT-III complex to the abscission site. Finally, we characterize a FIP3-endosome-dependent regulation ofthe ICB cortical actin network through the delivery of p50RhoGAP. These results provide a framework for the coordinated efforts ofactin, FIP3 endosomes and the ESCRTs to regulate cytokinesis and abscission.

Cytokinesis begins with a primary ingressionmediated by a contractionof an actomyosin ring1, leading to the formation of the ICB, whichis further resolved by thinning of the ICB from ∼2 µm to ∼100 nm(secondary ingression)2, followed by the ICB plasma membranefusion (abscission)2–4. Recent studies have shown that secondaryingression and abscission involve depolymerization of ICB actin andmicrotubules2–4, fusion of FIP3 endosomes2,5–7 and recruitment ofthe ESCRT complexes8–10. However, although it was shown thatmidbody accumulation of ALIX and TSG101 (ESCRT-I) recruitsCHMP4B (ESCRT-III) during late cytokinesis3,8,10, the roles of FIP3endosomes and ESCRT complexes during the formation of thesecondary ingression and abscission remain to be fully understood.Here, we demonstrate that secondary ingression is mediated by

FIP3-endosome fusion and occurs before ESCRT-III recruitment tothe abscission site, and that ESCRT-III is required only for stabilizationof the formed secondary ingression and final membrane scission.Furthermore, we use time-lapse microscopy and organelle proteomicsto identify secretory carrier membrane protein 2/3 (SCAMP2/3)and p50RhoGAP as membrane-traffic- and actin-regulating proteins

1Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA.2Department of Structural Biology and Biophysics, School of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA.3Department of Cell Biology and Physiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA. 4Department of Cell Biology, Schoolof Medicine, University of Virginia, Charlottesville, Virginia 22908, USA.5Correspondence should be addressed to R.P. (e-mail: [email protected])

Received 30 May 2012; accepted 9 August 2012; published online 23 September 2012; DOI: 10.1038/ncb2577

required for abscission, which are delivered to the ICB by FIP3endosomes. We support these findings by demonstrating that FIP3-endosome delivery of SCAMP2/3 or p50RhoGAP regulates actindepolymerization within the ICB and recruitment of ESCRT-IIIto the abscission site.

RESULTSESCRT-III recruitment to the midbody coincides with theaccumulation of FIP3 endosomesMany reports have shown that FIP3- and Rab11-positive endosomesaccumulate at the ICB and are required for abscission2,6,7,11,12, andthat the movement of FIP3 from centrosomes to the ICB marks theprogression of cells from early to late telophase (Supplementary Fig.S1A). A recent study suggested that endosomes are not required forabscission, on the basis of an observation that RAB8 endosomes arenot present at the ICB during late cytokinesis4. As it is unclear whetherRAB8 actually marks FIP3 endosomes, we compared RAB8 and FIP3during cytokinesis and show that there is little co-localization betweenRAB8 and FIP3 during early telophase, as RAB8 endosomes are

1068 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

∗ ∗

∗ ∗∗

∗∗

h

∗ ∗

b

d e g

a c

∗ ∗

∗∗ ∗

mCherry–CEP55 FIP3–GFP BF

∗ ∗

RFP–RAB8 FIP3–GFP Merge

RFP–RAB8 FIP3–GFP Merge

BF BFmCherry–CHMP4B

FIP3–GFPmCherry–TSG101

FIP3–GFPBFmCherry–CEP55

FIP3–GFP

f

BFmCherry–TSG101

FIP3–GFP BFmCherry–CHMP4B

FIP3–GFP

Figure 1 Spatiotemporal distribution of FIP3 endosomes, RAB8endosomes, CEP55 and ESCRT complexes during cytokinesis. (a–h)Fixed-cell imaging of HeLa FIP3–GFP cells co-expressing RFP–RAB8(a,b) and live-cell imaging of mCherry–CEP55 (c,d), mCherry–TSG101(e,f) and mCherry–CHMP4B (g,h). HeLa FIP3–GFP cells were imaged

in early cytokinesis (a,c,e,g) or late cytokinesis (b,d,f,h). The arrowsin b correspond to FIP3 and RAB8 co-localization outside theintercellular bridge. The outlined regions (d,f,h) mark enlarged areas.The asterisks mark the midbodies. BF, bright-field micrograph. Scalebars, 5 µm.

delivered to the ICB before FIP3-endosome accumulation (Fig. 1a).Subsequently, during late telophase, FIP3 endosomes occupy theICB, whereas RAB8 endosomes are no longer present within theICB (Fig. 1b). Interestingly, RAB8 and FIP3 co-localize on a smallpopulation of endosomes outside the ICB (Fig. 1b). This demonstratesthat during cytokinesis RAB8 and FIP3 mark different endocyticpopulations within the ICB (Fig. 1a,b).Various ESCRT complex components have been documented to

accumulate at the midbody, culminating in CHMP4B (ESCRT-III)recruitment during late telophase3,4,8,10. To establish the timing ofFIP3-endosome and ESCRT transport to the ICB, we co-imaged FIP3and ESCRT complex components CEP55, TSG101 and CHMP4Bduring cytokinesis. As previously reported3,8,13, CEP55 and TSG101arrive at the midbody during early telophase, with FIP3 endosomesstill residing outside the ICB (Fig. 1c–f). In contrast, CHMP4B arrivesat the midbody during late telophase (Fig. 1g–h), demonstratingthat ESCRT-III and FIP3 endosomes are both recruited to the ICB

during the final stages of cytokinesis, before the formation of thesecondary ingression.

CHMP4B is recruited to the abscission site after formation ofthe secondary ingressionPreviously it was suggested that FIP3 endosomesmediate the generationof the secondary ingression within the ICB before abscission2,12.Interestingly, two recent studies proposed that ESCRT-III oligomersalso play a role in the formation of the secondary ingression3,4. Tofurther define the involvement of FIP3 endosomes and ESCRT-IIIduring abscission, we used time-lapse microscopy to image theformation of the secondary ingression in HeLa cells co-expressingFIP3–GFP and mCherry–CHMP4B. We observed that the secondaryingression is able to form without CHMP4B moving to the siteof ICB narrowing (Figs 2a–e and 3a,b). Furthermore, FIP3–GFPfluorescence disappears during the formation of the secondaryingression (42 cells imaged; Fig. 2b,g). The disappearance of FIP3–GFP

NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 1069

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

a f g hBF

mCherry–CHMP4B

0 min 12 min 19 min 33 min

c

d

e

BF

mCherry–CHMP4B

∗0 min 38 min 68 min

b∗ ∗ ∗

∗FIP3–GFP

BF

∗ ∗

∗ ∗

∗ ∗

∗ ∗

FIP3–GFP

mCherry– CHMP4B

0 min

10 min

34 min

51 min

Figure 2 The secondary ingression forms before the recruitment ofCHMP4B. Mitotic HeLa FIP3–GFP cells expressing mCherry–CHMP4Bwere imaged using time-lapse microscopy. (a–e) Sequential time-lapseimages show abscission occurring either close (<2 µm; a–c) or far away

(>6 µm) from the midbody (d–e). (f–h) Sequential time-lapse images showfailed secondary ingression. In all images, the arrowheads mark secondaryingression and abscission sites. The asterisks mark the midbodies. Scalebars, 5 µm.

might be caused by the fusion of FIP3–GFP endosomes, or due toendosomes exiting the ICB. To investigate this, we imaged FIP3endosomes during mid-telophase (Supplementary Movie S1) orlate telophase (Supplementary Movie S2). Consistent with previousreports12,14, during mid-telophase FIP3 endosomes are activelydelivered to and from the ICB. In contrast, during late telophase,FIP3 endosomes are largely stationary and remain within the ICB.Together with previous reports2, this suggests that FIP3 endosomesfuse with the ICB plasma membrane during the formation of thesecondary ingression.This demonstrates that the formation of the secondary ingression is

not dependent on CHMP4B and probably is mediated by anothermechanism, possibly fusion of FIP3 endosomes. After secondaryingression, CHMP4B can be observed accumulating at the site of ICBthinning (Fig. 2c,e), marking the location where abscission occurs(5 cells imaged). Note, we never observed abscission without thedelivery of CHMP4B to the secondary ingression site. Furthermore, weoften see cases where secondary ingression was initiated, but was notfollowed by the recruitment of CHMP4B (Fig. 2f–h; >30 cells imaged).In these cases, the ICB regressed to its original width and did notundergo abscission (Fig. 2f–h), suggesting that whereas ESCRT-III isnot required for the initial formation of secondary ingression, it isrequired for the scission of the ICB.The factors that initiate ESCRT-III recruitment to the abscission site

remain elusive, but it was suggested that CHMP4B oligomers extendfrom the midbody to the abscission site3,4. In 27% of cells (6 of 22cells imaged) with CHMP4B at the abscission site, we observe possiblecontinuous CHMP4B spirals connecting the midbody to the abscissionsite (Supplementary Fig. S1G). However, in 73% of cells (16 of 22cells imaged), CHMP4B forms distinct puncta at the abscission site(Fig. 2c,e and Supplementary Figure 1H).Microtubule depolymerization within the ICB is required for

abscission1,3,4,14, and may determine the site of the secondaryingression2. To confirm that depolymerization of microtubules issufficient to cause secondary ingression, we imaged telophase cells

before and after the addition of nocodazole (7.5 µgml−1). Consistentwith the requirement of microtubule depolymerization for theformation of secondary ingression, the addition of nocodazole inducedrapid thinning of the ICB (Fig. 3d,e).As the secondary ingression forms before CHMP4B recruitment to

the abscission site (Fig. 2), we comparedmicrotubule depolymerizationwith FIP3 and CHMP4B recruitment to the abscission site.Consistent with previous work3,4, localized depolymerization ofmicrotubules (microtubule gaps) is observed in ∼40% of dividingHeLa cells (Fig. 3f–h and Supplementary Fig. S1B), with few ofthese sites containing CHMP4B (Fig. 3h and Supplementary Fig.S1C). In most cells, localized microtubule gaps did not containany detectable CHMP4B (Fig. 3f,g and Supplementary Fig. S1C),suggesting that ESCRT-III may not be absolutely required for localizedmicrotubule depolymerization. To investigate this possibility, wedepleted CHMP4B (Supplementary Fig. S1F) and analysed thepresence of microtubule gaps within the central spindle duringtelophase. As reported previously15,16, CHMP4B depletion increasedthe number of multinucleated cells (Supplementary Fig. S1D).Interestingly, CHMP4B knockdown had no effect on the formationof microtubule gaps (Supplementary Fig. S1E). Taken together,these results suggest that ESCRT-III is required only for the finalabscission step, and not microtubule depolymerization or secondaryingression formation.In addition to cells with microtubule gaps, some cells exhibited

depolymerization occurring at the minus ends of the microtubules(retracting microtubules) within the ICB (Fig. 3a,b and SupplementaryFig. S1B). In these cases, the formation of the secondary ingressionalways occurred behind the minus ends of the microtubules (Fig. 3a,b).Furthermore, kymographs (Fig. 3c) show increased ICB plasmamembrane dynamics behind the minus ends of the microtubules(Fig. 3c(A,B)), whereas the plasma membrane is stable where ICBmicrotubules are present (Fig. 3c(C)). Thus, microtubule clearingduring abscission may result from either of two processes—localizedsevering3,4 or minus-end retraction (Fig. 3a–c).

1070 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

BFBF

mCherry–CHMP4B mCherry–CHMP4B

∗ ∗ ∗ ∗

2.35 min

bd e

f

g

h

a

c

B

B

A

C

A C

–noc

∗∗∗

∗ ∗ ∗

∗ ∗ ∗

mCherry–CHMP4BAcetylated-tubulin Merge

BF

α-tubulin–GFP α-tubulin–GFP

–noc

BF α-tubulin–RFP α-tubulin–RFP

+noc +noc

3.8

μm

Figure 3 Clearance of microtubules from the ICB is requiredfor secondary ingression. (a–c) Mitotic HeLa cells expressingα-tubulin–GFP and mCherry–CHMP4B were analysed by time-lapsemicroscopy. The arrowheads mark the secondary ingression sites.The kymographs in c (from the cell shown in b) depict the ICBdynamics at the regions with (C) or without (A and B) detectablemicrotubules. (d,e) Mitotic HeLa cells expressing α-tubulin–GFP and

mCherry–CHMP4B were imaged before or after 10min incubationwith nocodazole (noc; 7.5 µgml−1). The arrowheads mark the pointwhere the secondary ingression site will be induced. (f–h) HeLacells expressing mCherry–CHMP4B were fixed and stained withanti-acetylated-tubulin antibodies. The arrowheads mark the regions ofmicrotubule severing/depletion. In all images (a–h), the asterisks markthe midbodies. Scale bars, 5 µm.

SCAMP2 and SCAMP3 are required for cytokinesisAs ESCRT-III and FIP3 endosomes accumulate at the secondaryingression site2, it raises a possibility that FIP3 endosomes deliverproteins that mediate the recruitment of ESCRT complexes. Recentwork determined that SCAMP3 interacts with TSG101 (refs 17–19). As SCAMPs reside in endosomes20, they potentially couldregulate ESCRT complex recruitment to the abscission site. Thus, weexamined the localization of different SCAMPs and have shown thatSCAMP1-3 localize to FIP3 endosomes during cytokinesis (Fig. 4a–gand Supplementary Fig. S2). Moreover, SCAMP1-3 localization at theICB is blocked after FIP3 knockdown (Fig. 4h–k and SupplementaryFig. S2). Knockdown of SCAMP2 or SCAMP3 caused an increase inthe number of multinuclear cells, whereas SCAMP1 depletion showedlittle effect (Fig. 5a,b). As SCAMP isoforms can have complementaryfunctions21–23, we also evaluated combinatorial knockdowns ofSCAMPs. Co-depletion of SCAMP2 and SCAMP3 resulted in anincrease in division failures (Fig. 5b), and in the time required forcells to complete cytokinesis (Fig. 5c–e).Originally SCAMPs were described as proteins that regulate

exocytotic membrane fusion21–23. To determine whether SCAMP2/3may regulate FIP3-endosomal fusion, we analysed the effect ofSCAMP2/3 knockdown on wave generation of the ICB plasmamembrane. It was previously shown that during late cytokinesis, theICB plasma membrane becomes very dynamic and that generationof the short-lived (few seconds) ICB plasma membrane protrusions,or waves24, occurs in a FIP3-endosome-dependent manner beforethe formation of the secondary ingression2. Consistently, SCAMP2/3knockdown resulted in a significant reduction in the number of

plasma membrane waves (Fig. 5f). Together, these results suggestthat SCAMP2/3 are required for abscission, possibly by regulatingFIP3-endosome fusion and/or mediating ESCRT-III assembly byrecruiting TSG101 to the abscission site.

FIP3 endosomes deliver p50RhoGAP to the ICB during latetelophaseInterestingly, the effect of SCAMP2/3 knockdown was not aspronounced as depletion of FIP3 (Fig. 5c,f), indicating that FIP3endosomes may also deliver other proteins that regulate cytokinesis.To identify these cargo proteins, we used proteomics to analyseimmuno-isolated FIP3 endosomes (Supplementary Fig. S3; seeMethods). This led to the identification of 441 proteins that arepresent only in immuno-isolated FIP3 endosomes (SupplementaryTable S1). Proteins known to bind ribosomes, mitochondria andDNA/RNA were eliminated as contaminants, leaving the remaining(103) proteins as candidates (Supplementary Fig. S3 and Table S1).Many of the candidate proteins have known roles in cytokinesis,but were not previously associated with FIP3 endosomes. We alsoidentified several proteins that were reported to reside in endosomes,but were never implicated in regulating cell division. One suchprotein was p50RhoGAP (also known as ARHGAP1), which has beenshown to reside in endosomes during interphase25,26. To confirmthat p50RhoGAP is present on FIP3 endosomes, we generated anti-p50RhoGAP antibody (Supplementary Fig. S4A–D) and have shownthat endogenous p50RhoGAP and FIP3 co-localize within the ICBduring late cytokinesis (Fig. 6a–f). FIP3 knockdown inhibited targetingof p50RhoGAP to the ICB, while having no effect on p50RhoGAP

NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 1071

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

a b c d

ih jFIP3–GFP BF

Merge

SCAMP2

SCAMP2

FIP3-KD

Mock MockEarly telophase Late telophase

k

Leve

ls o

f SC

AM

P2

in t

he IC

B

(arb

itrar

y flu

ores

cenc

e un

its p

er m

m2 )

Moc

k

FIP

3-K

D

∗∗∗ P < 0.0005

n =

11

n =

10

FIP3–GFP MergeSCAMP2

Merge

0

40

80

120

geFIP3–GFP

f

Figure 4 SCAMP1, 2 and 3 localize to the ICB during cytokinesisand their localization is dependent on FIP3 endosomes. (a–i) Mock-(a–g) or FIP3-siRNA-treated (h,i; FIP3-KD) HeLa FIP3–GFP cellswere fixed and stained with anti-SCAMP2 antibodies. The asterisksmark the midbodies. The image in g shows a magnification of the

area outlined in f. (k) Quantification of SCAMP2 localization withinthe ICB in mock-transfected or FIP3-siRNA-treated cells. The datashown are the means and s.d. of the anti-SCAMP2 fluorescenceintensity from 21 randomly chosen cells in telophase. Scale bars,5 µm (a–f,h–i) and 1 µm (g).

total cellular levels (Fig. 6g–j). Similarly, RFP-tagged p50RhoGAPco-localized with FIP3–GFP-containing organelles in live telophasecells (Supplementary Fig. S4E–H). Interestingly, we could also observe asmall pool of p50RhoGAP that is recruited to the midbody during earlytelophase and does not co-localize with FIP3 endosomes (Fig. 6b,c),suggesting that a small portion of p50RhoGAP can also be recruited tothe ICB in a FIP3-endosome-independent manner.To determine whether p50RhoGAP is required for cytokinesis, we

transfected cells with three different p50RhoGAP short interferingRNAs (siRNAs) to deplete endogenous p50RhoGAP (SupplementaryFig. S4A) and have shown that depletion of p50RhoGAP increasedthe number of multinucleate cells (Fig. 6k). To further confirm thatp50RhoGAP is required for cytokinesis, we analysed the divisionof p50RhoGAP-depleted cells by time-lapse microscopy. Althoughp50RhoGAP knockdown did not have any effect on the formationand initial ingression of the cleavage furrow, depletion of p50RhoGAPincreased the time required for cells to complete cytokinesis (Fig. 5c–e),and also increased the number of cells that failed abscission(Fig. 5e). FIP3 knockdown again had a much more pronouncedeffect on cytokinesis than p50RhoGAP knockdown (Fig. 5c–e andSupplementary Fig. S4A), suggesting that FIP3 endosomes functionby delivering multiple cytokinesis regulators, which include, but areprobably not limited to, p50RhoGAP and SCAMP2/3.

p50RhoGAP-dependent depolymerization of actin within theICB is required for abscissionAlthough actin cytoskeleton plays a key role in the progressionof the cleavage furrow27 and maintenance of cell shape duringcytokinesis28, depolymerization of the ICB cortical actin during latetelophase is required for successful completion of cytokinesis. Thisraises a possibility that FIP3 endosomes may deliver p50RhoGAPto the ICB to inactivate Rac/Rho GTPases and induce cortical actindisassembly. To investigate this, we imaged filamentous actin (F-actin)in dividing cells using a previously characterized F-actin-bindingprotein utrophin–GFP (ref. 29; Supplementary Fig. S5). In earlyanaphase to early telophase, F-actin is enriched at the cleavage furrow(Supplementary Fig. S5A–C). Similarly, GFP-tagged myosin IIA isalso enriched at the furrow during its formation and initial primaryingression (Supplementary Fig. S5D). However, on completion ofprimary ingression, myosin-IIA–GFP is no longer detectable withinthe ICB (Supplementary Fig. S5D). During late telophase, F-actin isalso gradually removed from the ICB (Supplementary Fig. S5A,C)and is no longer detectable within the forming secondary ingression(Supplementary Fig. S5B). To determine whether FIP3 endosomes arerequired for the ICB actin depolymerization, we imaged F-actin withinthe ICB using utrophin–RFP after knockdown of either SCAMP2/3or p50RhoGAP. Mock-transfected cells in late cytokinesis exhibit

1072 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

Mo

ck

SC

1-K

D#1

SC

2-K

D#1

SC

3-K

D#1

Mul

tinuc

leat

e ce

lls

(per

cent

age

of

tota

l)

Div

isio

n p

heno

typ

e (p

erce

ntag

e o

f to

tal)

n =

494

P < 0.001

∗∗ ∗∗

∗∗

∗∗

∗ P < 0.05

∗∗ P < 0.005

P <

0.0

1

P <

0.0

01

- SC1

- SC2

- SC3

- TfR

Moc

k

SC2/3-

KD#1

P < 0.001

n = 10

Mock

SC2/3-KD#1

2.5 min

Mock

BF

Division time (h)

Per

cent

age

of

com

ple

ted

d

ivis

ion

Div

isio

n tim

e (h

)

∗n

= 2

85

n =

377

n =

42

n =

24n

= 9

n =

17

n =

24

n =

24

n =

39

n =

364

n =

461

n =

463

n =

405

- SC1

- SC2

- SC3M

ock

Mo

ck

SC

2/3-

KD

#2

SC

2/3-

KD

#1

Moc

k

SC1-KD#1

SC2-KD#1

SC3-KD#1

SC1/2-

KD#1

SC2/3-

KD#2

SC2/3-

KD#1

CompletedConnected

FailedApoptosis

BF

SC2/3-KD#1

f

Moc

k

FIP3-

KD#1

FIP3-

KD#2

SC2/3-

KD#1

SC2/3-

KD#2

p50-

KD#2

p50-

KD#3

Moc

k

FIP3-

KD#1

FIP3-

KD#2

SC2/3-

KD#1

SC2/3-

KD#2

p50-

KD#2

p50-

KD#3

∗∗

∗∗

∗∗ P < 0.01

0

5

10

15

20

25

30

0102030405060708090

100

0 2 4 6 8 10 12 14 16 18

0

2

4

6

8

10

12

14

16

0102030405060708090

100

Num

ber

of

wav

es p

er

time-

lap

se s

erie

s (2

.5 m

in)

0

5

10

15

n = 7

ed

a b c

Mock (n = 42)

FIP3-KD#1 (n = 26)FIP3-KD#2 (n = 27)

SC2/3-KD#1 (n = 25)SC2/3-KD#2 (n = 28)p50-KD#2 (n = 46)

p50-KD#3 (n = 42)

Figure 5 SCAMP2 and SCAMP3 are required for successful completionof cytokinesis. (a) Western blot analysis probed for the indicated proteinsin single and double SCAMP knockdowns (KD). (b) Quantification ofmultinucleate HeLa cells in single or double SCAMP knockdowns. The datashown are means and s.d. from three independent experiments. n indicatesthe number of cells counted. The asterisks indicate a statistically significantdifference from the mock-transfected cells. (c–e) Mock, FIP3, SCAMP2/3or p50RhoGAP siRNA-treated (KD) HeLa cells were imaged by time-lapsemicroscopy for 24 h (time-lapse 1 h). The data shown are the percentageof cells that completed division (c), the average time required for cells to

divide (d) and the division phenotype (e). n indicates the number of cellscounted. The error bars represent s.d. of the mean. The asterisks indicate astatistically significant difference from the mock-transfected cells. (f) Mock-or SCAMP2/3-siRNA-treated cells were imaged by time-lapse microscopyto analyse the dynamics of the ICB during late telophase, as determinedby FIP3–GFP localization within the ICB (images on right). Shown arerepresentative kymographs used to quantify the dynamics of the ICB. Thehistogram depicts means and s.d. of the data. n is the number of randomlychosen cells in late cytokinesis. Scale bars, 5 µm. Uncropped images ofblots are shown in Supplementary Fig. S7.

little cortical F-actin within the ICB, as compared with the cell body(Supplementary Fig. S6A,D). In contrast, SCAMP2/3 or p50RhoGAPknockdown resulted in significant increases in the total cortical F-actinlevels within the ICB (Supplementary Fig. S6B–D).To further confirm that FIP3, SCAMP2/3 and p50RhoGAP are

required for actin disassembly at the ICB, we also visualized F-actinin fixed HeLa cells with rhodamine–phalloidin. We analysed thelevels of F-actin during anaphase and telophase, and consistent withthe utrophin–RFP data, early telophase F-actin was enriched at theICB (Fig. 7a–f). In contrast, during late telophase (as determinedby the presence of FIP3 endosomes within the ICB), the levels ofF-actin were significantly decreased (Fig. 7g–j). The depletion ofSCAMP2/3, p50RhoGAP or FIP3 using siRNAs led to statisticallysignificant increases in the F-actin levels at the ICB during latetelophase (Fig. 7k–s). These data imply that FIP3-endosome-dependentdelivery of SCAMP2/3 and p50RhoGAP to the ICB is required for thedepolymerization of the cortical F-actin network before abscission.

SCAMP2/3 and p50RhoGAP are required for the recruitment ofCHMP4B to the abscission siteOur data demonstrate that FIP3, SCAMP2/3 and p50RhoGAP are re-quired for actin depolymerization within the ICB, and as disassembly ofthe cortical acto-myosin network is probably required for the thinningof the ICB, we investigated whether SCAMP2/3 and p50RhoGAP arealso required for the formation of the secondary ingression. We usedbright-field time-lapse microscopy to image secondary ingressions inlate telophase cells (n= 91), and over 45% had a clearly identifiablestable secondary ingression (Fig. 8a,c and Supplementary Movie S3).Depletion of p50RhoGAP or SCAMP2/3 resulted in a decrease in thefrequency of the secondary ingression (Fig. 8c). In contrast, CHMP4Bknockdown had no effect on the ability of cells to form the secondaryingression (Fig. 8b,c and Supplementary Movies S4 and S5). Thus, con-sistent with our previous observations, p50RhoGAP and SCAMP2/3 arerequired for the initial formation of the secondary ingression, whereasCHMP4B is probably required only for final scission of the ICB.

NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 1073

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

Mul

tinuc

leat

e ce

lls

(per

cent

age

of

tota

l)

P < 0.01

Early telophase

Late telophase

Late telophase

p50RhoGAP Merge

BF

FIP3–GFP

b c

FIP3-KD

MockP < 0.001

Leve

ls o

f p

50R

hoG

AP

in t

he IC

B

(arb

itrar

y flu

ore

scen

ce u

nits

per

mm

2 )

n = 11

-FIP1

-FIP3

Mo

ck

FIP

3-K

D

-p50

P < 0.001

P < 0.01

Mergep50RhoGAPFIP3–GFP

Mock

FIP3–GFP p50RhoGAP

n = 9

0

40

80

120

160

Mo

ck

p50

-KD

#1

p50

-KD

#2

p50

-KD

#3

n =

365

n =

341

n =

396

n =

743

0

4

8

12

16

h i

e f

a

g

d

Mock FIP3-KD

j

k

Figure 6 Localization of p50RhoGAP during cytokinesis. (a–f) DividingHeLa cells expressing FIP3–GFP were stained with anti-p50RhoGAPantibody. (g–i) FIP3 was depleted by RNA-mediated interference (KD) andcells were stained with anti-p50RhoGAP antibodies. The asterisks markthe midbodies. Scale bars, 5 µm. (j) Quantification of p50RhoGAP levelsat the ICB in mock- or FIP3-siRNA-treated cells. The data shown are the

means and s.d. of 20 randomly picked cells in late telophase. (k) Cellstransduced with three different p50RhoGAP siRNAs were analysed fordefects in cytokinesis by counting the number of multinucleate cells. Thedata shown are the means and s.d. of three independent experiments. n isthe total number of cells counted. Uncropped images of blots are shownin Supplementary Fig. S7.

Although CHMP4B is recruited to the abscission site during thefinal stages of cytokinesis, it remains unclear whether the formationof this secondary ingression induces ESCRT-III recruitment. It hasbeen shown that the ESCRT-III complex preferentially associateswith >100 nm diameter high-curvature membranes, a diametersimilar to that of the secondary ingression30. Furthermore, itwas previously shown that the SCAMP3 associates with TSG101(ref. 17), a protein known to recruit the ESCRT-III complex. Todetermine whether FIP3 endosomes are required for ESCRT-IIIrecruitment, we analysed the localization of CHMP4B–YFP duringlate telophase after FIP3, SCAMP2/3 or p50RhoGAP knockdown. Inmost cells, CHMP4B formed a double ring flanking the midbody(Fig. 8d), which was previously described3,4. Only in ∼25% ofcells, were we able to detect CHMP4B present at the secondaryingression sites, suggesting that these cells are at the final stageof abscission (Fig. 8e,f). In FIP3-, SCAMP2/3- or p50RhoGAP-knockdown cells we could observe a significant decrease in thenumber of cells with CHMP4B located at the abscission site(Fig. 8f). As a whole, the data indicate that FIP3-endosome-dependent delivery of SCAMP2/3, p50RhoGAP and probably otherregulatory proteins is required for the ICB actin depolymerization,the formation of the secondary ingression and the subsequenttranslocation of CHMP4B from the midbody to the abscission sitewithin the ICB.

DISCUSSIONRecent studies have identified FIP3 endosomes, actin, microtubulesand ESCRT complexes as regulators of the abscission. However,the exact mechanisms and linkages between these players remainelusive. Here, we show that FIP3 endosomes have an essentialrole in the formation of the secondary ingression, placementof the ESCRT-III machinery and regulating actin cytoskeletondisassembly within the ICB.It has been established that depolymerization of microtubules

within the ICB is a key step in allowing the final separation ofdaughter cells, and that the microtubule-severing enzyme spastinplays an important role in localized clearing of microtubulesat the future abscission site4,31. This study also identifies apreviously uncharacterized alternative mechanism of microtubuledepolymerization during cytokinesis by way of minus-endmicrotubuledepolymerization, which occurs away from midbody-associatedESCRT-III. This mechanism seems to be more common in cellsforming long ICBs and may explain why this mechanism wasnot observed in MDCK cells, where ICBs are less extended3. Itis likely that the mechanism of microtubule clearance is cell typedependent. For example, epithelial cells may have a higher dependenceon spastin-mediated severing3, and highly motile cells, such asfibroblasts, could rely on minus-end depolymerization. This couldalso explain why spastin knockdown only delays, but does not

1074 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

Furr

ow a

ctin

(arb

itrar

y flu

ores

cenc

e un

its)∗

BF

P < 0.05P < 0.01

∗∗∗∗

∗∗∗∗

∗∗

∗∗

∗ ∗

∗ ∗ ∗

∗ ∗ ∗

a b c s

g

k

o BF

Actin Actin Actin

Actin

p50-KD#2

SC2/3-KD#1

Mock

Actin FIP3

Mid-telo

Late-telo

Late-telo

Early-teloAnaphase

∗∗∗∗

n = 11

Actin FIP3

Actin FIP3

Actin FIP3

Actin FIP3

Actin

Actin

Late-telo

Actin FIP3

BF

h

l

p

i

m

r

j

n

q

d e f

Moc

k

p50-K

D#2

p50-K

D#3

SC2/3-

KD#1

SC2/3-

KD#2

FIP3-

KD#1

FIP3-

KD#2

n = 13

n = 13n = 12

n = 12n = 12

n = 9

0

150

100

50

Figure 7 FIP3-mediated p50RhoGAP and SCAMP2/3 deliveryis required for actin depolymerization during late telophase.(a–s) Mock- (a–j,s), p50RhoGAP- (k–n,s), SCAMP2/3- (o–q,s) orFIP3- (s) siRNA-treated HeLa cells were fixed and stained withrhodamine–phalloidin. a,d show a cell in anaphase. b,e show a cellin early telophase. c,f show a cell in mid-telophase. g–q show cellsin late telophase, as determined by the presence of FIP3–GFP in the

ICB. j,n and q are high-magnification images of the areas outlined ing,k and o. (s) The quantification of rhodamine–phalloidin fluorescenceintensity within the ICB during late telophase. The data shown are themeans and s.d. n indicates the number of randomly chosen cells. Thearrows in j,n and q point to the region within the ICB that was usedfor quantification of the rhodamine–phalloidin fluorescence intensity.Scale bars, 5 µm

block, abscission2,31. Thus, the combination of several microtubuledepolymerization mechanisms probably contributes to clearing theICB of microtubules during abscission.The sequential building of the ESCRT machinery in the midbody

has been well documented3,8,10. Although the involvement of ESCRTcomplexes in cytokinesis is clear, how this fits with the role of FIP3endosomes remains undetermined. Here, we compare spatiotemporaldynamics of secondary ingression formation, ESCRT recruitmentand FIP3-endosome delivery. We show that FIP3 endosomes arepresent throughout the entire ICB, including the site of secondaryingression. Although time-lapse analysis confirmed that CHMP4Bis required for ICB membrane scission, the data also show thatCHMP4B is recruited to the abscission site after the formation ofthe secondary ingression. Furthermore, CHMP4B depletion does nothave any effect on microtubule gap formation or the initiation of thesecondary ingression, suggesting that the ESCRT complexes do notmediate the formation of the secondary ingression. A question arisesof what regulates the movement of CHMP4B from the midbody tothe site of the secondary ingression. Recent data using yeast ESCRT

complexes found that ESCRT-II and CHMP6 act together to directESCRT-III polymerization to regions of highly curved membranes invitro, with a preference towards >100 nm liposomes30. Consistent withthat, ESCRT-dependent intraluminal multivesicular body generationand viral budding32–37 generate vesicles ranging in size from 50 to75 nm, well below the 2 µm diameter of the ICB. On the basis ofour findings, we propose that the FIP3-endosome-induced secondaryingression is required to establish a region of highly curved membrane,thus enabling the recruitment of ESCRT-III. The formation of thesecondary ingression before the arrival of CHMP4B also alleviatespotential size restrictions placed on the configuration of ESCRTmachinery during scission.Another barrier for ESCRT-mediated abscission is the presence of

a plasma-membrane-associated cortical actin network. This corticalF-actin network serves to structurally form, maintain and hold theshape of the ICB during early telophase. Recently, the importanceof the actin–myosin cortical network in stabilizing and positioningthe cleavage furrow during cytokinesis has been demonstrated38. Thisactin cortical network needs to be disassembled for abscission to take

NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 1075

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

Secondary ingression

Abscission

Midbody

1–1.5 μm

0.1 μm

Microtubule severing/endosome fusionActin depolymerization

ESCRT-III recruitment to the abscission site

Actin network

Microtubules

Furrow endosomes

ESCRT-III

SCAMP2/3

∗∗

∗∗ ∗∗ ∗∗

CH

MP

4B a

t th

e ab

scss

ion

site

(p

erce

ntag

e of

tot

al in

tel

opha

se)

Moc

k

FIP3-

KD

p50-K

D#2

p50-K

D#3

SC2/3-

KD#1

d

c

CHMP4B tubulin∗∗

e

∗∗

n =

68

n =

70

n =

65

n =

103

n =

97

n =

79

Cel

ls w

ith s

econ

dar

y in

gres

sion

(p

erce

ntag

e of

tot

al in

late

tel

opha

se)

P < 0.01P < 0.005

∗∗

∗∗

P < 0.005P < 0.05

∗∗∗

∗ ∗∗

n =

91

n =

70

n =

50

n =

68

n =

55

n =

45

a

b

CHMP4B-KD

Mock

FIP3–GFP

FIP3–GFP

Moc

k

p50-K

D#2

p50-K

D#3

SC2/3-

KD#1

SC2/3-

KD#2

CHMP4B

-KD

CHMP4B

CHMP4B CHMP4B tubulin

SC2/3-

KD#20

10

20

30f

Midbody

Midbody

Midbody

Midbody

g

0

20

40

60

Figure 8 FIP3-mediated SCAMP2/3 and p50RhoGAP delivery recruitsCHMP4B to the secondary ingression and abscission site. (a–c) HeLacells expressing FIP3–GFP were treated with p50RhoGAP, SCAMP2/3 orCHMP4B siRNAs (KD). The presence or absence of the stable secondaryingression was then determined by time-lapse imaging of cells in telophase.The asterisks mark the midbody; the arrowheads mark secondary ingression.(c) Quantification of the number of telophase cells with secondary ingression.The data shown are means and s.d. derived from three separate experiments.n indicates the number of telophase cells counted. The asterisks indicate a

statistically significant difference from the mock. (d–f) HeLa cells expressingYFP–CHMP4B were treated with p50RhoGAP, SCAMP2/3 or FIP3 siRNAs.Cell were then fixed and stained for acetylated tubulin. The asterisks markthe midbody; the arrowheads mark the microtubule gap/abscission site.(f) Quantification of the number of telophase cells with CHMP4B at theabscission site. The data shown are means and s.d. derived from threeseparate experiments. n indicates the number of telophase cells counted.The asterisks indicate a statistically significant difference from the mock.(g) Proposed abscission model (see text for details). Scale bars, 5 µm.

place. Consistently, a recent study shows that the inhibition of ICBactin disassembly during late cytokinesis results in abscission defects39.Here we also observe that the formation of the secondary ingressionis preceded by the depolymerization of the cortical actin withinthe ICB. The identification of several Rac/Rho and actin regulatorswithin immuno-isolated FIP3 endosomes in our proteomic studies

raised an interesting possibility that FIP3 endosomes may regulate thedepolymerization of the cortical actin network. Consistent with that,inhibition of FIP3-endosome fusion by SCAMP2/3 knockdowns alsoblocked actin disassembly within the ICB and inhibited cytokinesis.In addition, knockdown of the FIP3-endosome cargo protein,p50RhoGAP, also blocks actin disassembly within the ICB, inhibits

1076 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

CHMP4B recruitment to the secondary ingression sites and leads to latecytokinesis defects. Similarly, recent work39 demonstrated that Rab35endosomes also regulate actin disassembly within the ICB by regulatinglevels of phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2). AsRab35 seems tomark endosomes that are distinct fromFIP3 endosomes,there may be at least two independent endocytic pathways of actinregulation, Rab11 or Rab35 dependent40.Here we show that SCAMP2/3 may be functioning to regulate

FIP3-endosome fusion, as signified by the inhibitory effects ofknockdown on membrane dynamics. However, it seems that theSCAMP roles also manifest post-fusion, assisting in organizingCHMP4B to the abscission site. Although the exact mechanisms ofSCAMP2/3 recruitment of CHMP4B are unclear, SCAMP3 mighthelp organize ESCRT assembly at the abscission site by way ofbinding to TSG101 through its PSAP motif17. The interactionof SCAMPs with phosphoinositides41 may control PtdIns(4,5)P2

distribution in a manner that promotes the ESCRT-III recruitmentand actin depolymerization. Indeed, SCAMP2/3 knockdown inhibitsthe recruitment of CHMP4B to the microtubule gaps, the putativefuture abscission sites.In summary we propose that FIP3 endosomes regulate abscission

by delivering actin-regulating and ESCRT-III recruitment proteins tothe ICB. The fusion of these endosomes with the plasma membraneresults in localized depolymerization of the cortical actin cytoskeleton,leading to the formation of the secondary ingression (Fig. 8c). Thethinning of the ICB to less than 100 nm, as well as delivery of theTSG101-binding protein SCAMP3, then allows for the recruitmentof the ESCRT machinery, and abscission of daughter cells (Fig. 8c).Proteomic identification of other putative FIP3-endosome proteinscan now serve as a starting point to unravel FIP3-endosome controlof the complex ICB remodelling events that must occur to allow forabscission. �

METHODSMethods and any associated references are available in the onlineversion of the paper.

Note: Supplementary Information is available in the online version of the paper

ACKNOWLEDGEMENTSWe thank C. Pearson and C. Willenborg (University of Colorado Denver, USA)for critically reading the manuscript. We also thank J. Martin-Serrano (King’sCollege London, UK) for mCherry–CEP55, mCherry–TSG101 and YFP–CHMP4B;M. Geiszt (Semmelweis University, Hungary) for GFP–p50RhoGAP; C. Westlake(NCI-Frederick, USA) for RAB8 complementary DNA; A. Bresnick (Albert EinsteinCollege of Medicine, USA) for myosin-IIA–GFP; and W. Bement (University ofWisconsin Madison, USA) for utrophin–GFP and utrophin–RFP. This work wassupported in part by a grant from NIH-NIDDK (DK064380 to R.P.).

AUTHOR CONTRIBUTIONSJ.A.S. and R.P. conceived the project and wrote the manuscript with the help ofD.C. and C.C.W. Proteomic round one was performed and analysed by G.C.S.and J.W. Proteomic round two was performed and analysed by J.A.S. and J.W.Proteomic round three was performed and analysed by J.A.S. and C.Z. All remainingexperiments and data analysis were performed by J.A.S. and R.P.

COMPETING FINANCIAL INTERESTSThe authors declare no competing financial interests.

Published online at www.nature.com/doifinder/10.1038/ncb2577Reprints and permissions information is available online at www.nature.com/reprints

1. Eggert, U. S., Mitchison, T. J. & Field, C. M. Animal cytokinesis: from parts list tomechanisms. Annu. Rev. Biochem. 75, 543–566 (2006).

2. Schiel, J. A. et al. Endocytic membrane fusion and buckling-induced microtubulesevering mediate cell abscission. J. Cell Sci. 124, 1411–1424 (2011).

3. Elia, N., Sougrat, R., Spurlin, T. A., Hurley, J. H. & Lippincott-Schwartz, J.Dynamics of endosomal sorting complex required for transport (ESCRT) machineryduring cytokinesis and its role in abscission. Proc. Natl Acad. Sci. USA 108,4846–4851 (2011).

4. Guizetti, J. et al. Cortical constriction during abscission involves helices ofESCRT-III-dependent filaments. Science 331, 1616–1620 (2011).

5. Baluska, F., Menzel, D. & Barlow, P. W. Cytokinesis in plant and animal cells:endosomes ‘shut the door’. Dev. Biol. 294, 1–10 (2006).

6. Fielding, A. B. et al. Rab11-FIP3 and FIP4 interact with Arf6 and the exocyst tocontrol membrane traffic in cytokinesis. EMBO J. 24, 3389–3399 (2005).

7. Wilson, G. M. et al. The FIP3-Rab11 protein complex regulates recycling endosometargeting to the cleavage furrow during late cytokinesis. Mol. Biol. Cell 16,849–860 (2005).

8. Carlton, J. G., Agromayor, M. & Martin-Serrano, J. Differential requirements for Alixand ESCRT-III in cytokinesis and HIV-1 release. Proc. Natl Acad. Sci. USA 105,10541–10546 (2008).

9. Lindas, A. C., Karlsson, E. A., Lindgren, M. T., Ettema, T. J. & Bernander, R. Aunique cell division machinery in the Archaea. Proc. Natl Acad. Sci. USA 105,18942–18946 (2008).

10. Morita, E. et al. Human ESCRT and ALIX proteins interact with proteins of themidbody and function in cytokinesis. EMBO J. 26, 4215–4227 (2007).

11. Horgan, C. P., Walsh, M., Zurawski, T. H. & McCaffrey, M. W. Rab11-FIP3 localisesto a Rab11-positive pericentrosomal compartment during interphase and tothe cleavage furrow during cytokinesis. Biochem. Biophys. Res. Commun. 319,83–94 (2004).

12. Simon, G. C. et al. Sequential Cyk-4 binding to ECT2 and FIP3 regulatescleavage furrow ingression and abscission during cytokinesis. EMBO J. 27,1791–1803 (2008).

13. Gromley, A. et al. Centriolin anchoring of exocyst and SNARE complexes atthe midbody is required for secretory-vesicle-mediated abscission. Cell 123,75–87 (2005).

14. Montagnac, G. et al. ARF6 Interacts with JIP4 to control a motor switchmechanism regulating endosome traffic in cytokinesis. Curr. Biol. 19,184–195 (2009).

15. Carlton, J. G., Caballe, A., Agromayor, M., Kloc, M. & Martin-Serrano, J. ESCRT-IIIgoverns the Aurora B-mediated abscission checkpoint through CHMP4C. Science336, 220–225 (2012).

16. Morita, E. et al. Human ESCRT-III and VPS4 proteins are required forcentrosome and spindle maintenance. Proc. Natl Acad. Sci. USA 107,12889–12894 (2010).

17. Aoh, Q. L., Castle, A. M., Hubbard, C. H., Katsumata, O. & Castle, J. D. SCAMP3negatively regulates epidermal growth factor receptor degradation and promotesreceptor recycling. Mol. Biol. Cell 20, 1816–1832 (2009).

18. Falguieres, T., Castle, D. & Gruenberg, J. Regulation of the MVB pathway by SCAMP3.Traffic 13, 131–142 (2012).

19. Mota, L. J., Ramsden, A. E., Liu, M., Castle, J. D. & Holden, D. W. SCAMP3is a component of the Salmonella-induced tubular network and reveals aninteraction between bacterial effectors and post-Golgi trafficking. Cell. Microbiol.11, 1236–1253 (2009).

20. Castle, A. & Castle, D. Ubiquitously expressed secretory carrier membrane proteins(SCAMPs) 1–4 mark different pathways and exhibit limited constitutive traffickingto and from the cell surface. J. Cell Sci. 118, 3769–3780 (2005).

21. Liao, H. et al. Nonredundant function of secretory carrier membrane proteinisoforms in dense core vesicle exocytosis. Am. J. Physiol. Cell Physiol. 294,C797–C809 (2008).

22. Liu, L. et al. SCAMP2 interacts with Arf6 and phospholipase D1 and links theirfunction to exocytotic fusion pore formation in PC12 cells. Mol. Biol. Cell 16,4463–4472 (2005).

23. Zhang, J. & Castle, D. Regulation of fusion pore closure and compound exocytosisin neuroendocrine PC12 cells by SCAMP1. Traffic 12, 600–614 (2011).

24. Byers, B. & Abramson, D. H. Cytokinesis in HeLa: post-telophase delay andmicrotubule-associated motility. Protoplasma 66, 413–435 (1968).

25. Sirokmany, G. et al. Sec14 homology domain targets p50RhoGAP to endosomesand provides a link between Rab and Rho GTPases. J. Biol. Chem. 281,6096–6105 (2006).

26. Zhou, Y. T., Chew, L. L., Lin, S. C. & Low, B. C. The BNIP-2 and Cdc42GAPhomology (BCH) domain of p50RhoGAP/Cdc42GAP sequesters RhoA frominactivation by the adjacent GTPase-activating protein domain. Mol. Biol. Cell 21,3232–3246 (2010).

27. Pollard, T. D. Mechanics of cytokinesis in eukaryotes. Curr. Opin. Cell Biol. 22,50–56 (2010).

28. Robinson, D. N. & Cooley, L. Stable intercellular bridges in development: thecytoskeleton lining the tunnel. Trends Cell Biol. 6, 474–479 (1996).

NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012 1077

© 2012 Macmillan Publishers Limited. All rights reserved.

ART I C L E S

29. Burkel, B. M., von Dassow, G. & Bement, W. M. Versatile fluorescent probes for actinfilaments based on the actin-binding domain of utrophin. Cell Motil. Cytoskeleton64, 822–832 (2007).

30. Fyfe, I., Schuh, A. L., Edwardson, J. M. & Audhya, A. Association of the endosomalsorting complex ESCRT-II with the Vps20 subunit of ESCRT-III generates acurvature-sensitive complex capable of nucleating ESCRT-III filaments. J. Biol.Chem. 286, 34262–34270 (2011).

31. Connell, J. W., Lindon, C., Luzio, J. P. & Reid, E. Spastin couples microtubulesevering to membrane traffic in completion of cytokinesis and secretion. Traffic 10,42–56 (2009).

32. Demirov, D. G., Ono, A., Orenstein, J. M. & Freed, E. O. Overexpression of theN-terminal domain of TSG101 inhibits HIV-1 budding by blocking late domainfunction. Proc. Natl Acad. Sci. USA 99, 955–960 (2002).

33. Garrus, J. E. et al. TSG101 and the vacuolar protein sorting pathway are essentialfor HIV-1 budding. Cell 107, 55–65 (2001).

34. Martin-Serrano, J., Yarovoy, A., Perez-Caballero, D. & Bieniasz, P. D. Divergentretroviral late-budding domains recruit vacuolar protein sorting factors by usingalternative adaptor proteins. Proc. Natl Acad. Sci. USA 100, 12414–12419 (2003).

35. Martin-Serrano, J., Zang, T. & Bieniasz, P. D. Role of ESCRT-I in retroviral budding.J. Virol. 77, 4794–4804 (2003).

36. Nickerson, D. P., West, M., Henry, R. & Odorizzi, G. Regulators of Vps4 ATPaseactivity at endosomes differentially influence the size and rate of formation ofintralumenal vesicles. Mol. Biol. Cell 21, 1023–1032 (2010).

37. Von Schwedler, U. K. et al. The protein network of HIV budding. Cell 114,701–713 (2003).

38. Sedzinski, J. et al. Polar actomyosin contractility destabilizes the position of thecytokinetic furrow. Nature 476, 462–466 (2011).

39. Dambournet, D. et al. Rab35 GTPase and OCRL phosphatase remodel lipids andF-actin for successful cytokinesis. Nat. Cell Biol. 13, 981–988 (2011).

40. Kouranti, I., Sachse, M., Arouche, N., Goud, B. & Echard, A. Rab35 regulates anendocytic recycling pathway essential for the terminal steps of cytokinesis. Curr. Biol.16, 1719–1725 (2006).

41. Liao, H. et al. Secretory carrier membrane protein SCAMP2 and phosphatidylinositol4,5-bisphosphate interactions in the regulation of dense core vesicle exocytosis.Biochemistry 46, 10909–10920 (2007).

1078 NATURE CELL BIOLOGY VOLUME 14 | NUMBER 10 | OCTOBER 2012

© 2012 Macmillan Publishers Limited. All rights reserved.

DOI: 10.1038/ncb2577 METHODS

METHODSMaterials, antibodies and expression constructs. Reagents were obtained fromInvitrogen, Sigma-Aldrich, Clontech or Fisher, unless otherwise specified. Mousemonoclonal anti-HA (catalogue number sc-7392) antibody, used as per the man-ufacture’s recommendation, was purchased from Santa Cruz Biotechnology andrabbit anti-CHMP4B antibody (catalogue number ab105767), used as per the man-ufacture’s recommendation, was obtained from Abcam. Rabbit anti-GFP, SCAMP1(western blotting: 0.1 µgml−1; immunofluorescence analysis: 1 µgml−1), SCAMP2(serumused at 1:1,000 for western blotting, 1:500 for immunofluorescence analysis),SCAMP3 (western blotting: 0.1 µgml−1; immunofluorescence analysis: 1 µgml−1)and SCAMP4 (western blotting: 1 µgml−1; immunofluorescence analysis: 5 µgml−1)antibodies were previously described1,12,21,22. Rabbit polyclonal anti-p50RhoGAP an-tibodies were generated using recombinant purified GST–p50RhoGAP as previouslydescribed12 and used at 0.5 µgml−1. Secondary antibodies conjugated with Alexa488or Texas red were purchased from Jackson ImmunoResearch.

The following DNA constructs were previously described: FIP3–GFP (refs 6,7), mCherry–CEP55 (ref. 8), mCherry–TSG101 (ref. 8), YFP–CHMP4B (ref. 8)and p50RhoGAP–GFP (ref. 25). HA–TPD52 (ref. 42), pmEGFP–α-tubulin-C1(Addgene Plasmid 21039; ref. 43) and pmRFP–α-tubulin-C1 (Addgene Plasmid21040; ref. 43) were obtained from the Addgene plasmid repository. RFP–RAB8was a gift from C. Westlake. p50RhoGAP–GFP and p50RhoGAP-RFP weregifts from M. Geiszt. mCherry–CHMP4B was constructed by generating aPCR fragment of CHMP4B from CHMP4B–YFP using the forward primer5′-AAACTCGAGTCGGTGTTCGGGAAGCTGTTC-3′ and the reverse primer 5′-TTGAATTCTTACATGGATCCAGCCCAGTT-3′. The CHMP4B PCR fragmentand Clontech pmCherry-C1 plasmid were digested with Xho1 and EcoRI restrictionenzymes (New England Biolabs) and subsequently re-ligated.

Nocodazole drug treatment. All live HeLa cells were staged and imaged beforedrug treatment. Nocodazole, (Sigma-Aldrich) at a 7.5 µgml−1 final concentration,was added to live HeLa cells for 10min before obtaining images.

Light microscopy. For immunofluorescence microscopy, cells were fixed with4% paraformaldehyde, and then permeabilized in phosphate-buffered saline (PBS)containing 0.4% saponin, 0.2% BSA and 1% fetal bovine serum. Cells were stainedby standard immunofluorescence procedures and imaged with an inverted ZeissAxiovert 200M deconvolution microscope. Images were acquired and processedusing Intelligent Imaging Innovations three-dimensional rendering and explorationsoftware.

For time-lapse microscopy, cells were placed on collagen-coated 35mm glass bot-tomdishes (MatTek) for 24 h. Cells were thenmounted on a culture dish heater (DH-35; Warner Instruments) fitted with a TC-344B dual automatic temperature con-troller (Warner Instruments), and imaged at 37 ◦C using a×63 oil-immersion lens.

Fluorescent intensity data were determined either by a line scan through thefluorescent region or the total fluorescence within an outlined area by usingthe Intelligent Imaging Innovations three-dimensional rendering and explorationsoftware.

To determine the ratio of utrophin fluorescent intensity, a linewas drawn throughthe entire ICB, or a 5 µm line was drawn away from the cell body/ICB border alongthe plasma membrane. The data were then expressed as arbitrary fluorescence unitsper square micrometre and the ratio between the ICB and cell body fluorescenceintensity was calculated. Eight to twelve randomly chosen cells were analysed forevery condition.

Transfection and RNA interference. To knockdown FIP3, p50RhoGAP,SCAMP1, SCAMP2, SCAMP3 and VAMP8, HeLa cells were transfected with2 nM of specific siRNA using Lipofectamine 2000 (Invitrogen) accordingto the manufacturer’s protocol. Transfected cells were incubated for 48 h,trypsinized and plated on collagen-coated 35-mm glass-bottom dishes for24 h, and then processed for imaging or western blotting. siRNAs targetingSCAMPs (refs 17,21) (SCAMP1 siRNA1: 5′-UCAUCUCACUAGUUAAUGTT-3′; SCAMP2 siRNA1: 5′-CACUGUAGCCAACUUGCAUTT-3′, siRNA2: 5′-CGGACCCAGUGGAUGUAATT-3′; SCAMP3 siRNA1: 5′-CAGCUACUCGACAG-AACAATT-3′, siRNA2: 5′-AACGGAUCCACUCCUUAUATT-3′), FIP3 (siRNA1:5′-GGCGUGUGCUGGAGCUGGA-3′, siRNA2: 5′-GGCAGUGAGGCGGAGC-UGU-3′; ref. 7), p50RhoGAP (siRNA1: 5′-ACUUACAGGCCCACGCUCUTT-3′;ref. 25), and CHMP4B {Carlton, 2012 no. 203} (siRNA1: 5′-AUCGAUAAAGUUG-AUGAGUUATT-3′) were previously described. siRNAs targeting p50RhoGAP(siRNA2 and siRNA3) were designed by Thermo Scientific using the humanp50RhoGAP sequence (siRNA2: 5′-GGCGGAAGAUCAUUGUGUU-3′, andsiRNA3: 5′- UAACCUGGCUGUUGUUUUC-3′).

Immuno-isolation of FIP3–GFP endosomes from telophase cells. MagneticDyna-beads (Invitrogen) conjugated to protein A were crosslinked to either anti-rabbit IgG or anti-GFP, according to the manufacturer’s protocol. HeLa cells stablyexpressing FIP3–GFP were grown on 300 cm2 tissue culture flasks (Light Labs)and synchronized using a thymidine/nocodazole double block, and collected bymitotic shake off after a 90min incubation at 37 ◦C (Supplementary Fig. S3A).It was previously shown that under these conditions, most of these cells are inlate telophase12 (Supplementary Fig. S3B). Cells were pelleted and resuspended in10mM HEPES at pH 7.4. After a 10min incubation, the cells were mechanicallyhomogenized using a glass–glass homogenizer. The resulting lysate was centrifugedat 5,000g for 5min to remove cell debris. Salts were added to a final concentrationof 150mM NaCl and 1mM MgCl2 to the remaining microsomal preparation. Topre-clear the microsomal preparation, Dyna-beads conjugated to IgG were addedfor 1 h while rotating at room temperature. Pre-cleared microsomal preparationswere split into two different groups. For the control group, Dyna-beads conjugatedto rabbit IgG were added. For the sample group, Dyna-beads conjugated to rabbitpolyclonal anti-GFP antibody were added and incubated for 1 h while rotating atroom temperature. Beads fromeach groupwerewashed five timeswith 5ml of buffer(PBS with 10mM HEPES at pH 7.4, 150mM NaCl and 1mM MgCl2), and boundorganelles were eluted by 0.2M glycine at pH 2.5. After elution, the pH of the beadswas neutralized with 1M Tris at pH 11.

FIP3–GFP endosome proteomics. Control (IgG) and sample (FIP3–GFP)immuno-isolates were analysed in three independent proteomic studies (de-tails provided below). For each sample, the proteins were precipitated usingmethanol–chloroform44, solubilized using RapiGest (Waters, Corp.) and digestedwith trypsin (Promega).

In study 1, two aliquots of each IgG and FIP3–GFP digests were subjected tomicro liquid chromatography–tandem mass spectrometry45. Specifically, peptideswere separated with a microcapillary column (100 µm) using C18 reverse-phaseresin (5 µ, 125 Å, Aqua, Phenomenex) with a 2 h gradient (5%–65%). Tandemmassspectra obtained by data-dependent acquisition using a Thermo Electron LTQmassspectrometer were analysed using normalized Sequest46 and an IPI human proteindatabase (v3.45, 71983 sequences, released 10 June 2008) that had been concatenatedwith a randomized database. The resulting peptide identifications were assembledinto a protein list using DTASelect47 with requirements for at least two peptides perprotein, peptides that were fully digested by trypsin and at least 5 amino acids long.

In study 2, three aliquots of IgG and FIP3–GFP digest were subjected to microliquid chromatography–tandem mass spectrometry as in the first study. A newerversion of Sequest (v.2.7) and separate human protein and randomized databaseswere used to analyse the tandemmass spectra. Triplicate (IgG or FIP3–GFP) spectralmatches were evaluated using Percolator48. Unique peptide matches of at least 6amino acids with false discovery rate (q) values ≤0.01 were assembled into proteinlists using DTASelect with requirements for aminimumof two fully tryptic peptides.

In study 3, two immuno-isolates (IgG and FIP3–GFP telophase) were treatedand digested as above. Each sample was subjected to MudPIT analysis as previouslydescribed49. Specifically, peptides were separated with a microcapillary column(100 µm) using C18 reverse-phase resin (5 µ, 125 Å, Aqua, Phenomenex), andstrong cation exchange resin (Agilent Technologies) using eight increasing salt(ammonium acetate) pulses, each followed by a 2 h gradient (5%–65%). Tandemmass spectra obtained by data-dependent acquisition using a Thermo Electron LTQmass spectrometer were analysed as in the second study.

Only proteins identified in the anti-GFP antibody sample, but not the IgG control,were considered for further analysis (Supplementary Table S1). Proteins identifiedin both, IgG control and FIP3-endosome samples (separated by study), are listedin Supplementary Tables S2–S4. Multiple previously identified proteins that havebeen shown to associate with FIP3 endosomes and function during cytokinesis,namely centralspindlin50, dynein51, kinesin-1 (ref. 51), and VAMP8 (ref. 52), werealso identified in our studies, supporting the validity of our proteomic analysis.

Quantification. Multinucleation was counted by staining cells with Hoerscht andcounting the number of cells with an aberrant number of nuclei. At least 100randomly chosen cells were counted for every condition. The data shown are themeans and s.d. of at least three independent experiments.

To establish division times in cells treated with FIP3, p50RhoGAP or SCAMP2/3siRNAs, HeLa cells were plated on 35-mm glass-bottom dishes and 10 µm mini-stacks were acquired every hour for 24 h on an inverted Zeiss Axiovert 200Mmicroscope equippedwith an incubatorwith a heating stage environmental chamber(Pecon). The resulting images were analysed using ImageJ software (NIH) and thedivision time was calculated on the basis of cell morphology. At least 25 randomlychosen cells were used for analysis.

NATURE CELL BIOLOGY

© 2012 Macmillan Publishers Limited. All rights reserved.

METHODS DOI: 10.1038/ncb2577

To establish the frequency of CHMP4B–YFP localization to the abscission siteduring late telophase, the cells were treated with FIP3, p50RhoGAP or SCAMP2/3siRNAs. Cells were then fixed and stained with anti-acetylated-tubulin antibodies.Cells with CHMP4B–YFP present at the abscission site outside the midbodywere then counted. Only cells that were connected by an ICB and that hadCHMP4B–YFP at the midbody were analysed. Finally, only cells expressing lowlevels of CHMP4B–YFP were used for analysis. The experiment was repeated threetimes and the data shown are the means and s.d.

To determine the role of CHMP4B, SCAMP2/3 or p50RhoGAP on secondaryfurrow ingression, FIP3–GFP-expressing HeLa cells were treated with varioussiRNAs and plated on collaged-coated coverslips. At 24 h later, randomlychosen cells in late telophase (as determined by the presence of FIP3–GFPwithin the ICB) were analysed by bright-field time-lapse microscopy. To ensurethat we are not analysing short-lived plasma membrane waves (as describedin ref. 24), only cells that contained a stationary ICB thinning that wasmaintained during the entire time-lapse series (50 time points, total seriestime 5min) were scored as cells containing the secondary ingression. Theexperiment was repeated three times and the data shown are the meansand s.d.

To analyse p50RhoGAP or SCAMP2/3 levels within the ICB, cells were stainedwith anti-p50RhoGAP or anti-SCAMP antibodies. At least 20 (for each analysis)randomly picked cells in late telophase were imaged using the same exposure times(1.5 s for p50RhoGAP, 1 s for FIP3, 1.2 s for SCAMPs). The fluorescence intensitywithin the ICB was then analysed using Intelligent Imaging Innovations software.All data were expressed as arbitrary fluorescence units per square micrometre.

To analyse F-actin within the ICB, cells were either stained with rho-damine–phalloidin or transiently transduced with utrophin–RFP. At least ten (foreach analysis) randomly picked cells in late telophase were imaged using the sameexposure times. Only cells with a clearly defined ICB were used for this analysis. Thefluorescence intensity within the ICB was then analysed using Intelligent Imaging

Innovations software. All data were expressed as arbitrary fluorescence units persquare micrometre.

Statistical analysis was performed using GraphPad Prism using Student’s t -testwith a two-tailed, 95% confidence interval to generate P values.

42. Thomas, D. D., Frey, C. L., Messenger, S. W., August, B. K. & Groblewski, G. E. Arole for tumor protein TPD52 phosphorylation in endo-membrane trafficking duringcytokinesis. Biochem. Biophys. Res. Commun. 402, 583–587 (2010).

43. Steigemann, P. et al. Aurora B-mediated abscission checkpoint protects againsttetraploidization. Cell 136, 473–484 (2009).

44. Wessel, D. & Flugge, U. I. A method for the quantitative recovery of protein indilute solution in the presence of detergents and lipids. Anal. Biochem. 138,141–143 (1984).

45. McCormack, A. L. et al. Direct analysis and identification of proteins in mixturesby LC/MS/MS and database searching at the low-femtomole level. Anal. Chem. 69,767–776 (1997).

46. MacCoss, M. J., Wu, C. C. & Yates, J. R. 3rd Probability-based validation ofprotein identifications using a modified SEQUEST algorithm. Anal. Chem. 74,5593–5599 (2002).

47. Cociorva, D., Tabb, D. L. & Yates, J. R. Validation of tandem mass spectrometrydatabase search results using DTASelect. Curr. Protoc. Bioinformatics/editorialboard, Unit 13.4, http://dx.doi.org/10.1002/0471250953.bi1304s16 (2007).

48. Brosch, M., Yu, L., Hubbard, T. & Choudhary, J. Accurate and sensitive peptideidentification with Mascot Percolator. J. Proteome Res. 8, 3176–3181 (2009).

49. Lowenthal, M. S., Howell, K. E. & Wu, C. C. Proteomic analysis of the stacked Golgicomplex. Methods Mol. Biol. 432, 37–49 (2008).

50. Glotzer, M. The molecular requirements for cytokinesis. Science 307,1735–1739 (2005).

51. Glotzer, M. The 3Ms of central spindle assembly: microtubules, motors and MAPs.Nat. Rev. Mol. Cell Biol. 10, 9–20 (2009).

52. Low, S. H. et al. Syntaxin 2 and endobrevin are required for the terminal step ofcytokinesis in mammalian cells. Dev. Cell 4, 753–759 (2003).

NATURE CELL BIOLOGY

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 1

DOI: 10.1038/ncb2577Supplemental Figure 1

A

Early Telophase Late Telophase

FIP3-GFP

MT

with

out

gap

s

MT

with

ga

ps

Min

us-e

ndre

tract

ion

B

0

40

80

20

60

n=102

M

icro

tubu

les

with

in th

e IC

B(p

erce

nt o

f tot

al c

ells

in te

loph

ase) C

0

40

80

20

60

n=63

CH

MP

4B lo

caliz

atio

n w

ithin

ICB

(per

cent

of t

otal

cel

ls w

ith M

T G

ap)

Mid

body

Onl

y

Mid

body

+MT

gap

D E

0

20

10

15

5

n=35

1

n=29

3

Moc

k

CH

MP

4B-K

D

Mul

tinuc

leat

e ce

lls (%

tota

l) p<0.01

Moc

k

CH

MP

4B-K

D

n=10

8

n=16

7

0

40

20

60

M

icro

tubu

les

with

gap

(per

cent

of t

otal

cel

ls in

telo

phas

e)

Moc

k

CH

MP

4B-K

D

F

-FIP3

-CHMP4B

G H

**

Figure S1 FIP3 and microtubule dynamics during late telophase. (A) Schematic representation of FIP3 dynamics during early and late telophase. (B) Quantification of telophase cells with microtubule (MT) gaps and minus-end retractions. The data shown are the means and standard deviation from three independent experiments. (C) Quantification of CHMP4B localization within the ICB in telophase cells. The data shown are the means and standard deviation from three independent experiments. (D-E) Quantification

of multinucleation (D) and microtubule gap formation (E) in cells treated with CHMP4B siRNA. The data shown are the means and standard deviation from three independent experiments. (F) Immunoblot analysis of lysates from mock or CHMP4B siRNA-treated HeLa cells. (G and H) Images of telophase Hela cells with CHMP4B-YFP extending from the midbody to the abscission site in “continuous” manner (G) or forming a distinct pool (H). Scale bars 5 μm.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

2 WWW.NATURE.COM/NATURECELLBIOLOGY

Supplemental Figure 2

A B C

D E F

G H E

J K L

Mock

Mock

FIP3-KD

FIP3-KD

SCAMP1

SCAMP1

SCAMP3

SCAMP3

Merge

Merge

BF

BF

FIP3-GFP

FIP3-GFP

FIP3-GFP

FIP3-GFP

Lev

els

of S

CA

MP

1 in

the

ICB

(arb

itrar

y flu

ores

cenc

e un

its/m

m2)

Moc

k

FIP

3-K

D

0

40

80

100

20

60p<0.0005

n=10

n=10

* **

Moc

k

FIP

3-K

D

p<0.0005

n=10

n=10

0

40

80

100

20

60

120

140

Lev

els

of S

CA

MP

3 in

the

ICB

(arb

itrar

y flu

ores

cenc

e un

its/m

m2)M

* **

N

FIP3-GFP

FIP3-GFP

SCAMP3

SCAMP2

FIP3-GFP SCAMP1

SC3-KD

SC2-KD

SC1-KD

* *

* *

* *

Figure S2 Localization of SCAMP1 and SCAMP3 during telophase. (A-L) Mock (A-C, G-E) or FIP3 siRNA-treated (D-F, J-L) HeLa FIP3-GFP cells were fixed and stained with either anti-SCAMP1 (A and D) or anti-SCAMP3 (G-J) antibodies. Asterisk marks midbodies. (M) Quantification of SCAMP1 or SCAMP3 immunofluorescence within the ICB. The data shown are the

means and standard deviations of fluorescence from 10 randomly picked cells in telphase. (N) HeLa FIP3-GFP cells were treated with either SCAMP1, SCAMP2 or SCAMP3 siRNAs and stained with anti-SCAMP1 (top panels), anti-SCAMP2 (middle panels) or anti-SCAMP3 (bottom panels) antibodies. Scale bars 5 μm.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 3

Input α-IgG Input α-GFP

HeLa-FIP3-GFP HeLa-FIP3-GFP

Synchronize (Thymidine/Nocodazole)

Microsomes

Immunoprecipitation

anti-GFPIgG

Mass-Spec Analysis

A B

C

59% cells in telophase

FIP3-GFP

BF

*

*

*

*

*

*

#

#

-FIP3-GFP

-FIP1

-FIP1

-Syntaxin6

-TfR

Supplemental Figure 3

Total Identified Proteins (441)DNA/RNA associated 35.1%Misc 24.9%Mitochondria associated 7.7%ER proteins 5.7%Unknown 3.2%Candidate proteins 23.4%

Membrane transport 6.3%Actin regulators 5.4%Kinases & phosphatases 3.9%Lipid regulators 2.7%Tubulin regulators 2.5%Motor 1.8%Intermediate filament associated 0.7%

D

Figure S3 Immunoisolation and proteomic analysis of FIP3-GFP endosomes. (A) HeLa FIP3-GFP cells were synchronized using thymidine/nocodazole block. Cells were isolated by mitotic shake off, washed and incubated for 90 minutes. Cells were then lysed using glass-glass homogenizer and microsomes isolated. FIP3-endosomes were then immunoisolated using rabbit polyclonal anti-GFP antibody. Rabbit IgG was used as control. (B) HeLa FIP3-GFP cells were imaged 90 minutes after release from thymidine/nocodazole block. Asterisks mark cells in late

telophase. Pound sign marks cell in metaphase. (C) Western blot analysis FIP3-GFP enrichment within immunoisolated FIP3-endosome. Microsomes (input), IgG and anti-GFP immunoisolates were separated by SDS/PAGE and immunoblotted with anti-GFP, anti-FIP1, anti-TfR and anti-synatxin6 antibodies. TfR and FIP1 are the markers of recycling endosomes that are not enriched within the ICB. Syntaxin 6 is a marker for TGN. (D) Proteins identified in immunoisolated FIP3-endosomes were categorized based on their function. Scale bars 5 μm.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

4 WWW.NATURE.COM/NATURECELLBIOLOGY

Supplemental Figure 4

E F GFIP3-GFP Mergep50RhoGAP-RFP

H

FIP

3-G

FPp5

0Rho

GA

P-R

FP

*

**

*

*

**

*

*

**

*

*

**

*

* **

*

* **

*

-p50RhoGAP

-Transferrin Receptor

Moc

k

p50-

KD

#2

p50-

KD

#1

p50-

KD

#3A

KD levels57.18% 93.11% 94.34%

B C DFIP3-GFP FIP3-GFP FIP3-GFP

Mock Mock p50-KD#2

p50RhoGAP Ab p50RhoGAP Abp50RhoGAP AbGST-p50RhoGAP

Moc

k

FIP

3-K

D#2

FIP

3-K

D#1

-FIP3

-FIP1

KD levels62.9% 56.9%

5 um

5 um

Figure S4 Time-lapse analysis of p50RhoGAP-RFP dynamics during late telophase. (A) Western blot analysis of p50RhoGAP and FIP3 levels in cells treated with either p50RhoGAP or FIP3 siRNAs. Transferrin receptor and FIP1 are used as loading controls. (B-D) Mock (B and C) or p50RhoGAP siRNA-treated (D) FIP3-GFP expressing HeLa cells were stained using rabbit anti-p50RhoGAP antibody in the presence (C) or absence (B and D) of 10-

fold excess of purified recombinant GST-p50RhoGAP protein. FIP3-GFP was also imaged to identify cell in late telophase. (E-H) HeLa cells co-expressing FIP3-GFP and p50RhoGAP-RFP were analyzed by time lapse microscopy. Square box in (E) marks the region that is enlarged in (H). In (H) panels asterisks mark the organelles that contain both, FIP3-GFP and p50RhoGAP-RFP. Scale bars 5 μm (B-D), 10 μm (E-G), and 1 μm (H).

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 5

Furr

ow a

ctin

/Bod

y ac

tin

(a

rbitr

ary

valu

es)

D

A

0 min 4 min 13 min 1 hr 55 min

Ut-GFP

B Ut-GFPBF

**

0

1

2

3

4

5

0 min 8 min

11 min 1 hr 24 min

MyoIIA-GFP

**

C

1.05

2.19

4.31

0.80

Supplemental Figure 5

Figure S5 Filamentous actin is depolymerised in the ICB prior to abscission. Time-lapse images of HeLa cells expressing utrophin-GFP (A and B) or myosin IIA (D) were taken at the indicated time points. Lines in

A mark the 5μm region used for quantification of f-actin levels (C). Arrows mark the secondary ingression sites. Asterisks mark the midbody. Scale bars 5 μm.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

6 WWW.NATURE.COM/NATURECELLBIOLOGY

A

B BF

BF Ut-RFP

Ut-RFP FIP3-GFP

FIP3-GFP

Furr

ow a

ctin

/Bod

y ac

tin

(arb

itrar

y un

its)

D

Mock

SC2/3-KD#1

*

*

p<0.05U

t Flu

ores

cenc

e (a

rbitr

ary

units

)

C BF Ut-RFP FIP3-GFP

p50-KD#2

* * *

Supplemental Figure 6

*

*

*

*

Ut F

luor

esce

nce

(arb

itrar

y un

its)

Ut F

luor

esce

nce

(arb

itrar

y un

its)

Furrow actinBody actin

Mock

SC2/3-K

D#1

SC2/3-K

D#2

p50-K

D#2

p50-K

D#30.0

0.5

1.0

1.5

2.0

2.5

3.0

p<0.0001

*

**

**

****

n=22

n=12

n=6 n=10n=9

Figure S6 p50RhoGAP and SCAMP2/3 are required for actin depolymerization during late telophase. Mock (A), SCAMP2/3 siRNA-treated (B), or p50RhoGAP siRNA-treated (C) HeLa FIP3-GFP cells expressing utrophin-RFP were analyzed by live cell imaging. Only cells in late cytokinesis (as determined by FIP3-GFP localization within the ICB) were chosen for analysis. Asterisks mark the midbodies. Line through the ICB

indicates where line intensity scans were obtained (graphs on the right). (D) Ratiometric quantification of f-actin intensity within the ICB. (Inset) Graphic representation of the regions used for ratiometric quantification; black lines (furrow actin intensity) and grey lines (cell body actin). Data shown in D are the means and standard deviations derived from 8-20 randomly chosen cells in late telophase. Scale bars 5 μm.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 7

Supplemental Figure 7

Figure 5A-Left

Figure 5A-Right Figure 6J

Supplemental Figure 1F

Supplemental Figure 3C

Supplemental Figure 4A-Left Supplemental Figure 4A-Right

anti-SCAMP1

anti-p50RhoGAP

anti-GFP (FIP3-GFP)

anti-GFP (FIP3-GFP)

anti-SCAMP2

anti-FIP1

anti-FIP1

anti-SCAMP3

anti-Syntaxin6

anti-TfR

anti-TfR

anti-TfR

anti-CHMP4B anti-FIP3

anti-SCAMP2 anti-SCAMP3anti-SCAMP1 anti-FIP3 anti-FIP1 anti-p50RhoGAP

250-150-100-

50-

37-

50-

37-50-

37-

250-150-100-

50-

37-

250-150-100-

50-

37-

250-150-100-

50-

37-

250-150-100-

50-

37-

250-150-100-

50-

37-

M M M

250-150-100-

250-150-100-

50-

37-

50-

37-

M250-150-100-

250-150-100-

50-50-

37-37-

M MM

M

MMMM

250-

150-

100-

50-

250-

150-

100-

50-

250-

150-

100-

50-

50-

37-50-37-

50-37-

250-150-100-

50-

37-

150-100-

50-

37-

Figure S7 Full scans of all immunoblots shown in this study.

© 2012 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

8 WWW.NATURE.COM/NATURECELLBIOLOGY

Supplementary Tables

Supplemental Table 1 Putative FIP3-GFP endosome cargo proteins. Proteins were categorized based upon protein function analysis. Only proteins identified in immunoisolated FIP3-endosomes but not IgG controls are listed in this table. The shown list includes proteins identified in three different experiments.

Supplemental Table 2 List of all proteins identified in IgG control and anti-GFP sample from experimental run #1.

Supplemental Table 3 List of all proteins identified in IgG control and anti-GFP sample from experimental run #2.

Supplemental Table 4 List of all proteins identified in IgG control and anti-GFP sample from experimental run #3.

Supplemental Movie 1 FIP3-GFP endosome movement during mid-telophase.FIP3-GFP expressing HeLa cell at mid-telophase was imaged by time-lapse microscopy. Shown move consists of 50 sequential images. Exposure 200 ms, time-lapse 500 ms. Cells were determined to be in mid-telophase based on the fact that FIP3-GFP has started to accumulate at the ICB, with the large pool of FIP3-endosomes still present at the cell bodies. Shown cell is a representative of 25 imaged cells.

Supplementary Movies

Supplemental Movie 2 FIP3-GFP endosome movement during late-telophase.FIP3-GFP expressing HeLa cell at late-telophase was imaged by time-lapse microscopy. Shown move consists of 50 sequential images. Exposure 200 ms, time-lapse 500 ms. Cells were determined to be in late-telophase based on the fact that FIP3-GFP endosomes are now present only in the ICB. Shown cell is a representative of 25 imaged cells.

Supplemental Movie 3 Formation of the secondary ingression in mock-treated HeLa cell.Late telophase HeLa cell with secondary ingression and expressing FIP3-GFP was imaged by bright-field time-lapse microscopy. Shown move consists of 50 sequential images. Exposure 30 ms, time-lapse 5 sec.

Supplemental Movie 4 Formation of the secondary ingression in CHMP4B siRNA-treated HeLa cell.CHMP4B siRNA-treated late telophase HeLa cell with secondary ingression and expressing FIP3-GFP was imaged by bright-field time-lapse microscopy. Shown move consists of 50 sequential images. Exposure 30 ms, time-lapse 5 sec. FIP3-GFP time-lapse sequence of the same cell is shown in supplemental movie 5.

Supplemental Movie 5 FIP3-GFP dynamics during the formation of the secondary ingression in CHMP4B siRNA-treated HeLa cell.CHMP4B siRNA-treated late telophase HeLa cell with secondary ingression and expressing FIP3-GFP was imaged by time-lapse microscopy. Shown move consists of 50 sequential images. Exposure 200 ms, time-lapse 5 sec. Bright-field time-lapse sequence of the same cell is shown in supplemental movie 4.

© 2012 Macmillan Publishers Limited. All rights reserved.