ArticlePDF Available

Endosomal recycling controls plasma membrane area during mitosis

Authors:

Abstract and Figures

The shape and total surface of a cell and its daughters change during mitosis. Many cells round up during prophase and metaphase and reacquire their extended and flattened shape during cytokinesis. How does the total area of plasma membrane change to accommodate these morphological changes and by what mechanism is control of total membrane area achieved? Using single-cell imaging methods, we have found that the amount of plasma membrane in attached cells in culture decreases at the beginning of mitosis and recovers rapidly by the end. Clathrin-based endocytosis is normal throughout all phases of cell division, whereas recycling of internalized membranes back to the cell surface slows considerably during the rounding up period and resumes at the time at which recovery of cell membrane begins. Interference with either one of these processes by genetic or chemical means impairs cell division. The total cell-membrane area recovers even in the absence of a functional Golgi apparatus, which would be needed for export of newly synthesized membrane lipids and proteins. We propose a mechanism by which modulation of endosomal recycling controls cell area and surface expression of membrane-bound proteins during cell division. • clathrin • endocytosis • exocytosis • cell division • dynasore
Exocytosis is required during cell division. (A) Surface distribution of transferrin receptor and Lamp-1 during interphase and cytokinesis. HeLa cells were processed for immunofluorescence at 4°C by incubation with antibodies specific for the luminal domain of the transferrin receptor (green) and of Lamp-1 (red), followed by fixation and addition of fluorescently tagged secondary antibodies (Alexa-647 and Alexa-594, respectively). Images acquired in the absence (control) or presence of overexpressed cytosolic forms of the EGFP-labeled v-soluble N-ethylmaleimide-sensitive factor attachment protein receptors (V-SNAREs) (VAMP3-DN or VAMP7-DN). The cells expressing these constructs were identified by the cytosol EGFP signal (data not shown). The control panel (cytokinesis) corresponds to two optical sections 2 m apart. Cells in interphase contain little Lamp-1 at their cell surface and stain weakly for transferrin receptor. Some blebs present during cytokinesis (control) score positive for Lamp-1, whereas others are labeled with transferrin receptor; the overall transferrin receptor signal is significantly stronger when compared with cells in interphase. Short expression (4-6 h) of VAMP3-DN prevents the surface expression of transferrin receptor but not of Lamp-1 in cells undergoing telophase; in contrast, similar expression of VAMP7-DN prevents the surface appearance of Lamp-1 but not of transferrin receptor. DNA was labeled with DAPI (blue). Scale bar, 20 m. (B) Normal function of VAMP3 and VAMP7 is required for completion of cytokinesis. Top panel (control): Images acquired by bright field illumination of a BSC1 cell starting with anaphase, continuing through cytokinesis and ending with the spreading and separation of the two daughter cells. Middle panel (VAMP3-DN): images (from SI Movie 11) of BSC1 cell expressing VAMP3-DN for 4-6 h before imaging; blebs are present, ingression of the cleavage furrow occurs but cells do not separate. Bottom panel (VAMP7-DN): images (from SI Movie 12) of BSC1 cell expressing VAMP7-DN for 4-6 h before imaging; blebs are absent, ingression of the cleavage furrow occurs, but cells do not separate. Scale bar, 20 m.
… 
Content may be subject to copyright.
Endosomal recycling controls plasma membrane
area during mitosis
Emmanuel Boucrot and Tomas Kirchhausen*
Department of Cell Biology and CBR Institute for Biomedical Research, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115
Communicated by Marc W. Kirschner, Harvard Medical School, Boston, MA, March 16, 2007 (received for review November 21, 2006)
The shape and total surface of a cell and its daughters change
during mitosis. Many cells round up during prophase and meta-
phase and reacquire their extended and flattened shape during
cytokinesis. How does the total area of plasma membrane change
to accommodate these morphological changes and by what mech-
anism is control of total membrane area achieved? Using single-cell
imaging methods, we have found that the amount of plasma
membrane in attached cells in culture decreases at the beginning
of mitosis and recovers rapidly by the end. Clathrin-based endo-
cytosis is normal throughout all phases of cell division, whereas
recycling of internalized membranes back to the cell surface slows
considerably during the rounding up period and resumes at the
time at which recovery of cell membrane begins. Interference with
either one of these processes by genetic or chemical means impairs
cell division. The total cell-membrane area recovers even in the
absence of a functional Golgi apparatus, which would be needed
for export of newly synthesized membrane lipids and proteins. We
propose a mechanism by which modulation of endosomal recycling
controls cell area and surface expression of membrane-bound
proteins during cell division.
clathrin endocytosis exocytosis cell division dynasore
Acharacteristic of eukaryotic cells, particularly when spread
on a substrate and unprotected by a cell wall, is to round up
during mitosis, becoming more compact at metaphase and
recovering during cytokinesis. By altering its size and shape, the
cell presumably acquires a mechanism for modulating spatially
segregated signaling pathways as it divides (1) and for ensuring
transfer of a similar complement of constituents to the daughter
cells. Control of membrane dynamics must therefore be linked
to progress through the stages of cell division. Simple geometric
considerations argue that transformation from an extended cell
interphase to a rounded mitotic cell should be accompanied by
a large reduction in cell surface, particularly if the cellular
volume remains approximately constant after having doubled
during the S to M phase transition (2).
One way to accommodate the apparent change in cell area
during mitosis is to maintain the total amount of membrane at
the cell surface by allowing formation of extensive folds. Imaging
by scanning electron microscopy of the surface of synchronized
mastocytoma cells grown in suspension showed an increase in
the number of microvilli between G
1
and G
2
and an apparent
doubling of the surface area; based on these observations it was
proposed (3) that ‘‘cytokinesis is a physical unfolding or stretch-
ing process’’ that provides the extra surface required for the two
daughter cells. Similar studies done with adherent CHO or
BHK21 cells also showed a larger number of microvilli, blebs,
and ruff les in mitotic rounded cells than in the completely spread
cells in interphase (4, 5). In these early studies, however, it was
recognized that rapid membrane redistribution through endo-
cytosis and recycling rather than surface stretching could also
account for the changes in cell surface (4).
Another way to accommodate the altered surface-to-volume
ratio when cells round up is to regulate membrane traffic
between the cell surface and its interior, leading to membrane
loss at the onset of mitosis and recovery afterward. Imaging
along the cell cycle by electron microscopy of neuroblastoma
cells showed the appearance of blebs at the plasma membrane
and the accumulation of single-bilayer or multilamellar vesicles
close to the blebs (6). Based on these observations, it was
proposed that fusion of these structures with the plasma mem-
brane was responsible for a substantial increase in cell surface
area particularly at late stages during mitosis. Most recent
studies on the role of membrane traffic during cell division have
focused on delivery and retrieval of membrane at the cleavage
furrow during cytokinesis (for a recent review, see ref. 7). A
number of proteins found to be essential for cytokinesis also have
established functions in endocytic, exocytic, and recycling path-
ways. Interpretation of these findings have emphasized the role
of exocytic events during the actomyosin-based constriction
responsible for cleavage furrow formation and for abscission
(separation of the two daughter cells).
A number of observations demonstrate profound changes in
membrane traffic during mitosis. The Golgi apparatus disassem-
bles thus preventing constitutive exocytosis along the biosyn-
thetic pathway (8, 9). Moreover, transferrin uptake through the
clathrin-based endocytic pathway and f luid-phase uptake both
decline sharply during metaphase and anaphase (10–13) and
recover during telophase (14).
We have capitalized on newly introduced live-cell imaging
approaches to reinvestigate the role of membrane traffic in
regulating cell surface area. In particular, we have followed
individual cells through different stages of the cell cycle, instead
of monitoring bulk properties of synchronized populations of
cells. We have monitored changes in total plasma membrane in
cells undergoing mitosis, and we have correlated these data with
the dynamics of endocytosis and exocytosis during all stages of
cell division. Three key observations have emerged from these
studies: (i) the cell surface area decreases at the onset of mitosis,
when the mother cell rounds up, and recovers starting in
anaphase, with complete recovery before abscission; (ii) endo-
cytosis is normal throughout all phases of cell division; and (iii)
recycling of internalized membranes back to the cell surface
slows considerably during the rounding-up period and reacti-
vates by a concerted fusion of endosomes with the plasma
membrane, starting in anaphase. This recovery occurs even
under conditions in which the Golgi apparatus has been ren-
dered nonfunctional. We propose a simple mechanism in which
modulation of endosomal recycling controls surface area during
cell division.
Results
Cells Change Plasma Membrane Area During Mitosis. In preparation
for cell division, interphase cells growing on a substrate round up
Author contributions: E.B. and T.K. designed research; E.B. performed research; E.B.
analyzed data; and E.B. and T.K. wrote the paper.
The authors declare no conflict of interest.
Abbreviation: VAMP, vesicle associated membrane protein.
*To whom correspondence should be addressed. E-mail: kirchhausen@crystal.harvard.edu.
This article contains supporting information online at www.pnas.org/cgi/content/full/
0702511104/DC1.
© 2007 by The National Academy of Sciences of the USA
www.pnas.orgcgidoi10.1073pnas.0702511104 PNAS
May 8, 2007
vol. 104
no. 19
7939–7944
CELL BIOLOGY
(Fig. 1A), and their surface area, measured as described below,
decreases in a process that starts at the onset of prophase and
ends with metaphase (Fig. 1B). Reversal of these steps begins at
anaphase, continues through telophase, and becomes particu-
larly prominent during cytokinesis (Fig. 1 AC). Where does the
extra membrane go at the beginning of cell division, and where
does it come from at the end? To monitor with high temporal
resolution the total plasma-membrane area throughout a com-
plete cell-division cycle, we used three-dimensional, spinning
disk confocal, live-cell imaging of single cells labeled with the
membrane-impermeant dye, FM 1-43, which becomes f luores-
cent upon binding to the outer leaflet of the plasma membrane
(15) [Fig. 1Band supporting information (SI) Fig. 7A]. The
surface f luorescence signal is a measure of exposed membrane,
because the charged head group of FM 1-43 prevents it from
flipping to the inner leaflet, and internal membranes can only be
reached by endocytic vesicular traffic (15).
Cells selected at different stages during cell division were
incubated for a brief time with FM 1-43 at 37°C, and image stacks
were acquired after 2 min, a time sufficient to achieve a stable
fluorescent signal and short enough to reduce to a minimum the
contribution from dye association with endosomal membranes
because of endocytosis (Fig. 1B, z view). Any remaining intra-
cellular fluorescence was removed from each optical section by
manual masking (see Materials and Methods). The total surface
area within a population of cells at interphase varied widely (Fig.
1C). At metaphase, when cells rounded up, we found a large
reduction in that area, about two-fold for human HeLa cells and
about six- to eight-fold for monkey BSC1 cells (Fig. 1C).
Nonadherent human Jurkat T cells imaged in the same way
display a large amount of plasma-membrane projections during
interphase, in stark contrast to the fairly smooth appearance
during metaphase (SI Fig. 7A), suggesting that the amount of
plasma membrane is also lower during metaphase. As a com-
plementary approach to determine cell surface area, we moni-
tored the fluorescence signal at the plasma membrane elicited by
a chimera of EGFP fused at its C terminus with the peptide-
sorting motif CAAX expressed in BSC1 or HeLa cells (SI Fig.
7Band C). We followed the changes in cell surface of single cells
and found that it also increases during the transition from
metaphase to cytokinesis.
Cell rounding alone cannot account for the observed decrease
in surface area, because the surface remained constant in
interphase cells that rounded upon brief treatment with trypsin
(Fig. 1D). That is, the surface of trypsinized cells must ruffle or
fold when they round up. In addition, we ruled out a decrease in
FM 1-43 dye accessibility during cell rounding by observing that
the total amount of intracellular and plasma membrane available
(determined by total f luorescence) remained the same in mi-
totic, trypsinized, or interphase cells when we gently permeabil-
ized their plasma membrane with saponin before incubation w ith
the dye (SI Fig. 8A). These data confirm that the decrease of
plasma membrane during mitosis is matched by its accumulation
inside cells.
The compensatory increase in surface area began at the onset
of anaphase and continued throughout cytokinesis, until the two
daughter cells had recovered the total area originally present in
the mother cell (Fig. 1C). Similar results were obtained from
sequential measurements of surface area from the same cells
imaged at different stages during the cell cycle, using a modified
staining protocol (SI Fig. 8B). The period of cell membrane
reduction lasts 3045 min, the time it takes cells to round up and
reach metaphase. Recovery, which is relatively fast (15–20 min),
occurs before a perinuclear Golgi apparatus has assembled (SI
Fig. 8C).
Endocytosis Is Not Affected During Mitosis. It has been shown by
electron microscopy that cells contain plasma membrane coated
pits and vesicles regardless of their stage of cell division (16). All
endocytic plasma membrane coated pits and vesicles contain
clathrin adaptor AP-2 (17). We have now demonstrated that the
dynamics of AP-2 incorporation into plasma membrane pits and
vesicles during mitosis is the same as in interphase (Fig. 2). We
used BSC1 cells stably expressing
2-EGFP (17), part of AP-2
and imaged the surface attached to the coverslip to facilitate
data acquisition (Fig. 2 and SI Movies 1–3). Quantitative analysis
of these data (17) shows fluorescent
2-EGFP spots with
equivalent lifetimes during mitosis and interphase (SI Fig. 9 A
Fig. 1. Changes in cell shape and amount of plasma membrane during
mitosis. (A) BSC1 cells visualized at several stages during the cell cycle, using
bright field. N, nucleus. (B) BSC1 cells incubated for 2 min with FM 1-43 dye at
37°C and imaged at the same magnification at interphase, metaphase and
cytokinesis. Three-dimensional image stacks were obtained from sequential
optical sections acquired 0.25
m apart by using the spinning disk confocal
configuration. Shown is the fluorescence signal along the zaxis (Upper)atits
two dimensional projection (Lower). The integrated fluorescence, corrected
for any fluorescence signal inside the cells, represents the amount of plasma
membrane. Scale bar, 10
m. (C) Amount of plasma membrane at different
stages during the cell cycle was obtained in six experiments from 45 BSC1 and
64 HeLa cells, respectively. I, interphase; M, metaphase; C, cytokinesis. (D)
Amount of plasma membrane in BSC1 cells, rounded up immediately after
detachment by treatment with trypsin for 5 min at 37°C and then imaged as
in B. Data from two experiments from nine trypsinized cells and six untreated
control cells in interphase and spread on the coverslip.
7940
www.pnas.orgcgidoi10.1073pnas.0702511104 Boucrot and Kirchhausen
and B Left). Thus, the time needed to form a coated pit and for
it to bud as a coated vesicle is uniform throughout the cell cycle.
In addition, we could detect no change in the sizes of the coated
vesicles (SI Fig. 9B Right) or in the frequency of assembly events
per unit surface area (SI Fig. 9C). Similar results were obtained
with HeLa cells (SI Movie 4). These observations suggest that
endocytosis carried by the clathrin pathway remains normal
during mitosis.
Other forms of uptake probably also remain active during
mitosis, as the uptake of Alexa-594 labeled (10 kDa) dextran by
fluid phase endocytosis continued during mitosis (SI Fig. 10 A
and Bfor BSC1 cells and SI Fig. 10Cfor HeLa cells). As
expected, the internal membranes of rounding cells became
labeled by plasma membrane uptake when incubated steadily
with FM 1-43 during prophase and metaphase (SI Fig. 10D).
During mitosis, cells do not appear to internalize transferrin,
a ligand specifically taken up by clathrin-coated vesicles (12, 13).
These observations can be reconciled with our results, that
clathrin-based endocytosis remains uniformly active during the
cell cycle, if we recognize that significant reduction of recycling
would greatly decrease the amount of transferrin receptor
available on the cell surface. To test this hypothesis, we used
established methods (18, 19) to measure the uptake of f luores-
cently labeled transferrin and to normalize that uptake to the
number of accessible transferrin receptors with the potential to
internalize ligand (SI Fig. 11). The amount of transferrin bound
at the cell surface (Alexa488) or inside individual cells
(Alexa594) was calculated from two-dimensional projections of
three dimensional image stacks obtained by epif luorescence
microscopy at different stages during the cell cycle (SI Fig. 11 A
for BSC1 cells and SI Fig. 11Bfor HeLa cells). We observed a
substantial decrease in the amount of transferrin internalized
during prophase and metaphase (Fig. 3, Internalized and SI Fig.
11), consistent with all previous observations. We further ob-
served a marked decrease in the number of cell-sur face receptors
during prophase and metaphase followed by a very fast recovery
starting with anaphase, and becoming very prominent with
telophase and continuing into cytokinesis (Fig. 3 and SI Fig. 11,
Surface), also in agreement with previous obser vations (13). The
endocytic rate (18, 19), defined as the ratio of internalized
transferrin to transferrin bound at the cell surface (In/Sur ratio),
did not decrease during prophase and metaphase, but actually
showed a modest but significant increase compared with the rate
in cells during interphase or late stages of cytokinesis (Fig. 3).
Previous measurements were not done in a way in which this
ratio could have been measured in individual cells. Our results
show that the reduction in transferrin internalization during
mitosis does not correspond to an endocytic block, as formerly
concluded, but instead that most of the receptors remain trapped
within endosomes, most likely explained by a partial reduction in
recycling rates, which are then released back to the cell surface
as cytokinesis proceeds.
Recovery of the Plasma Membrane Correlates With Surface Bleb
Formation. Live cell imaging, using phase contrast or fluores-
cence microscopy of adherent cells incubated with FM 1-43
throughout the recovery phase, shows the appearance of a large
number of blebs at the outer surface of the cell, mostly at the
distal poles and away from the cleavage furrow (SI Fig. 12 and
SI Movies 5 and 6). These blebs appear and disappear rapidly
(seconds) (SI Fig. 12 A) until late stages of cytokinesis (SI Fig.
12B, 25 min). Bleb formation is not deleterious, because cells
that have completed cytokinesis spread and remain viable (SI
Fig. 12B, 52 min and SI Movie 6). Similar bebbling also occurs
in confluent HeLa cells (SI Fig. 12Cand SI Movie 7) and in the
nonadherent human Jurkat T and insect Sf9 cells (SI Fig. 12 D
and Eand SI Movies 8 and 9).
Recovery of the Plasma Membrane Does Not Require a Functional
Golgi Apparatus. The appearance of newly synthesized viral G
protein at the cell surface is strongly inhibited during metaphase,
anaphase, and early telophase (9). The images shown in SI Fig.
8Cindicate a diffuse intracellular distribution of the Golgi
marker, GalT-EGFP, during anaphase and telophase. Only
during cytokinesis does this marker start to appear punctate and
to localize in the perinuclear region (20, 21). Thus, a fully
reorganized Golgi apparatus is not required for recovery of
surface membrane. It is nonetheless possible that the substantial
exocytic traffic in anaphase and telophase requires a functional
postmitotic Golgi apparatus, even though a well localized GalT-
EGFP perinuclear signal is still absent (20, 21). We ruled out
involvement of a functional Golgi at this stage by observing the
appearance of blebs during anaphase and telophase and during
entrance to cytokinesis in cells treated with Brefeldin A before
entering mitosis (SI Fig. 12Fand SI Movie 10). The failure of
Brefeldin A to prevent the rapid recovery of plasma membrane
observed during anaphase and telophase shows that recovery
can occur without the contribution of newly synthesized mem-
branes from the Golgi. A similar Brefeldin A-insensitive mem-
brane deposition at the cleavage furrow of sea urchin zygotes has
been previously observed (22). However, a requirement for a
Fig. 2. Formation of clathrin coated pits and coated vesicles is not affected
by mitosis. Live cell fluorescence imaging of AP-2, containing clathrin coated
pits and coated vesicles located at the bottom surface of BSC1 cells. AP-2 was
labeled by stable expression of
2-adaptin fused to EGFP (17). Time series
collected for 6 min and at 37°C from one cell during interphase, and from
another cell sequentially imaged during metaphase and anaphase and 15
min into cytokinesis. The still images (Left) correspond to Middle and Bottom
optical sections (scale bars, 10 and 5
m, respectively) acquired after 3 min of
data collection; the kymographs (Right) represent the complete time-series.
The data are representative of experiments done in triplicate and were
acquired every 2 s with 1-s exposures, using the spinning disk confocal con-
figuration.
Fig. 3. Receptor-mediated endocytosis of transferrin during cell division.
The amounts of internalized transferrin (In) and of transferrin bound to the
cell surface (Sur) were obtained by integration of their corresponding fluo-
rescence signals present in the three-dimensional image stacks. The data
(mean standard deviation) were obtained from 24, 18, 14, 5, and 20 cells
imaged at interphase (I), prophase (P), metaphase (M), anaphase (A), and late
stages of cytokinesis (C), respectively.
Boucrot and Kirchhausen PNAS
May 8, 2007
vol. 104
no. 19
7941
CELL BIOLOGY
Brefeldin A-sensitive mechanism has been reported during
membrane deposition at the apex of the late cleavage furrow in
isolated, dividing C. elegans blastomeres (23) and during cell
plate during plant cytokinesis (24).
Exocytosis Controls the Recovery of Plasma Membrane. Some of the
blebs stain positively with an antibody specific for the luminal
domain of Lamp-1, a marker of late endosomes and lysosomes
normally absent from the cell surface during interphase (Fig.
4A, Interphase). Thus some blebs arise from fusion of Lamp-1
containing membranes with the plasma membrane (Fig. 4A,
Cytokinesis, control). Other blebs are labeled with the inter-
nalized transferrin receptor (Fig. 4A, Cytokinesis, control),
which accumulates even during mitosis in Lamp-1 negative
early/recycling endosomes (SI Fig. 13). Still other blebs have
neither Lamp-1 nor transferrin and may arise from additional
endosomal compartments. Presumably these are the multilayer
or single-membrane vesicles observed next to the blebs during
late mitosis (6). Formation of these blebs seems to require the
proper function of Myosin II, as their number was reduced in
cells incubated during telophase with blebbistatin, an inhibitor
of Myosin II (25) (SI Fig. 14A). Blebbistatin treatment also
decreased the reappearance of internalized Lamp-1, but barely
affected the recycling to the plasma membrane of transferrin
receptor (SI Fig. 14B). These observations suggest that during
cell division, the acto-myosin system has a role controlling
fusion, particularly of late endosomes/lysosomes with the
plasma membrane. They are consistent with the role of Myosin
II in cell membrane repair as mediated by calcium-dependent
exocytosis (26).
To further examine the relative contributions of early and late
endosomes to plasma-membrane recovery, we took advantage of
a published procedure (27, 28) to interfere with the fusion of
either type of endosome to the plasma membrane. VAMP3-DN
and VAMP7-DN are dominant negative cytosolic fragments of
v-soluble N-ethylmaleimide-sensitive factor attachment protein
receptors (V-SNAREs) specific for each path (27, 28). These
fragments were transiently overexpressed for 4 6 h in BSC1
cells, and those cells entering mitosis were selected for imaging.
We observed that expression of VAMP3-DN substantially de-
creased the reappearance of transferrin receptor (but not of
Lamp-1) at the cell surface following anaphase (Fig. 4Aand SI
Fig. 15A); this interference coincided with an almost complete
block in the recovery of plasma membrane (SI Fig. 15B) together
with a failure to undergo cytokinesis (Fig. 4Band SI Movie 11).
Expression for longer periods was not considered, because cell
division was strongly inhibited and was accompanied by multi-
ploidy in 80% of the cells (SI Fig. 15C). Likewise, short
expression of the corresponding cytosolic fragment of VAMP7
prevented Lamp-1 reappearance (but not of transferrin recep-
tor) at the cell surface (Fig. 4 Aand SI Fig. 15A), together with
a significant block in plasma membrane recovery (SI Fig. 15B).
Expression of VAMP7-DN also prevented cytokinesis (Fig. 4B
and SI Movie 12) and induced mutliploidy (SI Fig. 15D). We
conclude that both recycling and late endosomes participate in
membrane redeposition.
Clathrin-Mediated Endocytosis Is Required for Retrieval of Plasma
Membrane During Mitosis. Clathrin-mediated endocytosis is a
high capacity membrane traffic pathway. It generally carries at
least 50% of total endocytic traffic, and within 1 h it can
internalize the equivalent of the entire cell surface (29). If, as
proposed in Endocytosis Is Not Affected During Mitosis, endo-
cytosis remains active during all stages of cell division, whereas
exocytosis decreases substantially at the onset of mitosis, then
reducing endocytosis during mitosis should prevent the de-
crease in cell surface and retard or prevent mitotic roundup.
We confirmed this prediction in two ways. In one approach, we
reduced clathrin-based endocytosis by depleting, with RNA
interference in BSC1 (Fig. 5 and SI Fig. 16)orHeLa(SI Fig.
17) cells, the amount of
2-adaptin, a component required to
form the endocytic clathrin adaptor complex AP-2. As ex-
pected, 3 days of RNAi treatment was sufficient to block
transferrin uptake (Fig. 5, Overlay). All cells with an equato-
rial disposition of chromosomes (metaphase) failed to round
up; many (50%) had aberrant spindles (Fig. 5, DAPI and SI
Fig. 16A, quantification). All remained flat and large, and
their plasma membrane area was the same as control cells in
interphase with normal endocytosis (SI Fig. 16B). This treat-
ment also led to a significant accumulation of multinucleated
cells because of incomplete cell division (SI Fig. 16C). In a
second and complementary approach, we inhibited clathrin-
Fig. 4. Exocytosis is required during cell division. (A) Surface distribution of
transferrin receptor and Lamp-1 during interphase and cytokinesis. HeLa cells
were processed for immunofluorescence at 4°C by incubation with antibodies
specific for the luminal domain of the transferrin receptor (green) and of
Lamp-1 (red), followed by fixation and addition of fluorescently tagged
secondary antibodies (Alexa-647 and Alexa-594, respectively). Images ac-
quired in the absence (control) or presence of overexpressed cytosolic forms of
the EGFP-labeled v-soluble N-ethylmaleimide-sensitive factor attachment
protein receptors (V-SNAREs) (VAMP3-DN or VAMP7-DN). The cells expressing
these constructs were identified by the cytosol EGFP signal (data not shown).
The control panel (cytokinesis) corresponds to two optical sections 2
m apart.
Cells in interphase contain little Lamp-1 at their cell surface and stain weakly
for transferrin receptor. Some blebs present during cytokinesis (control) score
positive for Lamp-1, whereas others are labeled with transferrin receptor; the
overall transferrin receptor signal is significantly stronger when compared
with cells in interphase. Short expression (4 6 h) of VAMP3-DN prevents the
surface expression of transferrin receptor but not of Lamp-1 in cells under-
going telophase; in contrast, similar expression of VAMP7-DN prevents the
surface appearance of Lamp-1 but not of transferrin receptor. DNA was
labeled with DAPI (blue). Scale bar, 20
m. (B) Normal function of VAMP3 and
VAMP7 is required for completion of cytokinesis. Top panel (control): Images
acquired by bright field illumination of a BSC1 cell starting with anaphase,
continuing through cytokinesis and ending with the spreading and separation
of the two daughter cells. Middle panel (VAMP3-DN): images (from SI Movie
11) of BSC1 cell expressing VAMP3-DN for 4 6 h before imaging; blebs are
present, ingression of the cleavage furrow occurs but cells do not separate.
Bottom panel (VAMP7-DN): images (from SI Movie 12) of BSC1 cell expressing
VAMP7-DN for 4 6 h before imaging; blebs are absent, ingression of the
cleavage furrow occurs, but cells do not separate. Scale bar, 20
m.
7942
www.pnas.orgcgidoi10.1073pnas.0702511104 Boucrot and Kirchhausen
based endocytosis by acute interference with the function of
dynamin, a large GTPase essential for coated vesicle formation
(30–32). We incubated BSC1 cells for a brief interval with
dynasore, a recently discovered small molecule inhibitor of the
dynamin GTPase (33). Just as with the extended AP-2 deple-
tion, 30 min incubation with 80
M dynasore was sufficient to
block transferrin uptake, to prevent cell rounding (Fig. 5),
induce abnormal spindle formation (SI Fig. 16 A), and de-
crease cell surface area (SI Fig. 16B); 0.8% DMSO, used as
carrier, had no effect (Fig. 5).
Discussion
We have shown that the substantial decrease in total sur face area
that accompanies cell rounding is due mainly to a shutdown in
membrane recycling from endosomal compartments back to the
cell surface (Fig. 6). Most adherent cells in tissue culture round
when they divide, and cells within tissues also acquire a spherical
geometry during metaphase (34–37). This rounding appears to
be important for ensuing proper spindle formation and appro-
priate distribution of components to the daughter cells and for
establishing the correct intracellular spatial gradients for signal-
ing molecules (38). Indeed, we find that failure to round up,
induced by various forms of endocytic block, results in aberrant
spindle formation and incorrect cell division.
We have also shown that plasma membrane recovery is
essential for cell division. This rapid recovery is mediated by
massive fusion of endosomal membranes, starting at the onset
of anaphase and continuing through telophase. Our results
provide a satisfying explanation for recent observations ac-
quired in the course of RNAi screens aimed at identifying gene
products important for cell division (39 42). Clathrin heavy
chain, dynamin, Hsc70 (part of the uncoating machinery for
the clathrin coat), a number of plasma membrane and Golgi-
specific soluble N-ethylmaleimide-sensitive factor attachment
protein receptors (SNAREs), subunits of the Golgi COPI
complex, and regulatory small GTPases, such as Rab1 and
Rab7, are among theproteins whose depletion have a clear impact
in cell division.
The mechanisms that underlie membrane recovery studied
here are probably distinct from a number of other processes in
which new membrane is deposited. For example, specific struc-
tures such as the exocyst appear to participate in cleavage furrow
formation and abscission at cytokinesis (43), and Golgi-derived
vesicles are thought to contribute to the membranes laid down
during cellularization in Drosophila (44) and endocytic vesicles
(45). In plant cytokinesis, endocytic and Golgi-derived vesicles
are the sources of material for phragmoplast deposition (46). In
contrast, we propose here the formation of an internal mem-
brane reservoir during rounding up which stores membrane
components for subsequent release when called for later in cell
division.
In addition to the experiments described here, we have also
studied the potential role of Ca
2
in regulating exocytosis during
mitosis (see SI Results). We suggest that Ca
2
signaling is
essential for triggering the synchronous and coordinated fusion
with the plasma membrane of endomembranes stored inside the
cells, at a time when the Golgi apparatus has yet to assemble into
a fully functional organelle. A rise in intracellular Ca
2
triggers
rapid fusion of late endosomes and lysosomes with the plasma
membrane (see ref. 47 for recent review); a similar process, also
triggered by Ca
2
-influx during mechanical membrane injury,
governs wound healing. Similarly, the Ca
2
-dependent mem-
brane deposition required for the formation of a phagocytic cup
(48, 49) derives, at least in part, from VAMP3 dependent
recycling endosomes (27) and from VAMP7-dependent late
endosomes (50). Perhaps the use of these pathways during cell
division is the ancestral form. In contrast, normal traffic of
recycling endosomes is not known to depend on Ca
2
signaling,
at least during interphase.
It remains to be determined how modulation of these path-
ways occurs during mitosis. Control of these exocytic processes
must be linked to stages in cell division, to produce endosomal
recyling shutdown early in mitosis and to trigger abrupt reacti-
vation at anaphase.
Materials and Methods
Cell Preparation and Mitotic Stage Identification. The frequency of
adherent cells undergoing mitosis was increased from 1% to
10–20% by allowing them to reach 100% conf luency for 1 day,
a condition where a large fraction of the cells are arrested at the
end of G
1
(51). Cells were then trypsinized and seeded on 25-mm
diameter glass no. 1.5 coverslips at 50–70% conf luency; this
plating condition increases significantly the proportion of cells
simultaneously entering S phase at the time of seeding. Finally,
cells were imaged 12–14 h (HeLa) or 18–20 h (BSC1) after
Fig. 5. Endocytosis is required for retrieval of plasma membrane during
mitosis. BSC1 cells stably expressing EGFP-LCa were treated for 3 days with
RNAi for
2-adaptin to deplete AP-2 (center images) or for 30 min with 80
M
dynasore, a small molecule inhibitor of dynamin GTPase function (33) (right-
most image). These treatments inhibit clathrin-based endocytosis and during
mitosis prevent cell rounding and loss of cell membrane (see SI Fig. 16B). Cells
depleted of AP-2 and incubated for 5 min at 37°C with Alexa-594 transferrin
(red) display surface staining, an almost complete absence of internalized
transferrin, and the expected absence of endocytic clathrin coated pits and
vesicles (EGFP-LCa, green). Because of the relative brief incubation with
dynasore, these cells do not accumulate transferrin receptor at their surface
even though receptor endocytosis is blocked; the punctate pattern of EGFP-
LCa represents coated pits locked at the cell surface. A metaphase cell treated
with only 0.8% DMSO (Ctrl) is shown. Whereas most control mitotic cells have
normal spindles [decorated with EGFP-LCa, (53)], 50% of equivalent cells
depleted of AP-2 or treated with dynasore display aberrant spindles (see SI Fig.
16A). Scale bar, 20
m.
Fig. 6. Model for membrane traffic during different stages of the cell cycle.
The schematic representations highlight the changes in balance of membrane
traffic between endocytic routes and secretory together with recycling path-
ways during key stages of the cell cycle. These pathways are balanced during
interphase. During the rounding up occurring during prophase and meta-
phase, the amount of plasma membrane decreases because of the traffic
imbalance arising from normal endocytosis combined with a considerable
decrease in secretory and recycling traffic, creating an internal ‘‘membrane
reservoir.’’ During anaphase, telophase and cytokinesis the amount of plasma
membrane increases rapidly along with the appearance of surface blebs. Most
of the plasma membrane is recovered by the rapid fusion of the previously
stored endo-membranes with the cell surface. A Ca2signal is required to
trigger the rapid fusion of endomembranes with the plasma membrane in
what constitutes a form of regulated exocytosis. At this stage, the Golgi
apparatus is barely reassembled, and hence secretory traffic is still minimal.
Boucrot and Kirchhausen PNAS
May 8, 2007
vol. 104
no. 19
7943
CELL BIOLOGY
plating. Imaging by phase contrast bright field illumination was
used to determine the stage of single cells along the cell cycle
(selected from the unsynchronized population) according to the
following criteria: cells in interphase appear f lat, contain un-
condensed chromosomes surrounded by a nuclear envelope;
cells in prophase contain condensed chromosomes also sur-
rounded by a nuclear envelope; cells in metaphase appear round,
contain condensed chromosomes aligned at the metaphase plate
and lack their nuclear envelope; cells in anaphase appear less
round and contain two sets of condensed chromosomes each
starting to migrate toward opposite spindle poles; cells in
telophase start to develop the furrow or invagination separating
the two daughter cells and still contain condensed chromosomes
that reached the spindle poles; during cytokinesis, (imaged 20
min after the onset of anaphase), cells generate a deeper furrow,
still display condensed chromosomes while the nuclear envelope
starts to form around each chromosomal mass until abscission.
All live cell imaging were performed as described in ref. 52 and
SI Methods.
We thank Drs. A. Yu, S. Saffarian and R. Massol and the members of
our laboratory for their help and advice, and Dr. J. Stow and J. Luzio for
providing reagents. This work was supported by National Institutes of
Health Grants GM075252 and GM62566 (to T.K.), by the Perkin Fund
to purchase part of the imaging equipment used here, and the Interna-
tional Human Frontier Science Program Organization (to E.B.).
1. Meyers J, Craig J, Odde DJ (2006) Cur r Biol 16:1685–1693.
2. Ohnuma K, Yomo T, Asashima M, Kaneko K (2006) Bioorg Med Chem Cell Biol
7:25.
3. Knutton S, Sumner MC, Pasternak CA (1975) J Cell Biol 66:568–576.
4. Erickson CA, Trinkaus JP (1976) Exp Cell Res 99:375–384.
5. Porter K, Prescott D, Fr ye J (1973) J Cell Biol 57:815– 836.
6. Bluemink JG, van Maurik PA, Tertoolen LG, van der Saag P T, de Laat SW
(1983) Eur J Cell Biol 32:7–16.
7. Albertson R, Riggs B, Sullivan W (2005) Trends Cell Biol 15:92–101.
8. Lucocq JM, Warren G (1987) EMBO J 6:3239–3246.
9. Warren G, Featherstone C, Griffiths G, Burke B (1983) J Cell Biol 97:1623–
1628.
10. Berlin RD, Oliver JM, Walter RJ (1978) Cell 15:327–341.
11. Berlin RD, Oliver JM (1980) J Cell Biol 85:660– 671.
12. Sager PR, Brown PA, Berlin RD (1984) Cell 39:275–282.
13. Warren G, Davoust J, Cockcroft A (1984) EMBO J 3:2217–2225.
14. Schweitzer JK, Burke EE, Goodson HV, D’Souza-Schorey C (2005) J Biol
Chem 280:41628– 41635.
15. Brumback AC, Lieber JL, Angleson JK, Betz WJ (2004) Methods 33:287–
294.
16. Pypaert M, Lucocq JM, Warren G (1987) Eur J Cell Biol 45:23–29.
17. Ehrlich M, Boll W, Van Oijen A, Hariharan R, Chandran K, Nibert ML,
Kirchhausen T (2004) Cell 118:591–605.
18. Wiley HS, Cunningham DD (1982) J Biol Chem 257:4222–4229.
19. Ghosh RN, Gelman DL, Maxfield FR (1994) J Cell Sci 107(8):2177–2189.
20. Souter E, Pypaert M, Warren G (1993) J Cell Biol 122:533–540.
21. Jiang S, Rhee SW, Gleeson PA, Storrie B (2006) Mol Biol Cell 17:4105–4117.
22. Shuster CB, Burgess DR (2002) Proc Natl Acad Sci USA 99:3633–3638.
23. Skop AR, Bergmann D, Mohler WA, White JG (2001) Curr Biol 11:735–746.
24. Yasuhara H, Sonobe S, Shibaoka H (1995) Eur J Cell Biol 66:274–281.
25. Straight AF, Cheung A, Limouze J, Chen I, Westwood NJ, Sellers JR,
Mitchison TJ (2003) Science 299:1743–1747.
26. Togo T, Steinhardt R A (2004) Mol Biol Cell 15:688–695.
27. Murray RZ, Kay JG, Sangermani DG, Stow JL (2005) Science 310:1492–1495.
28. Martinez-Arca S, Alberts P, Zahraoui A, Louvard D, Galli T (2000) J Cell Biol
149:889–900.
29. Hansen SH, Sandv ig K, van Deurs B (1992) Exp Cell Res 199:19–28.
30. Praefcke GJ, McMahon HT, Engqvist-Goldstein AE, Drubin DG, Lang T,
Korolchuk V, Banting G, Song BD, Schmid SL, Wiejak J, et al. (2004) Nat Rev
Mol Cell Biol 5:133–147.
31. Schmid SL, McNiven MA, De Camilli P (1998) Curr Opin Cell Biol 10:504 –512.
32. McNiven MA (1998) Cell 94:151–154.
33. Macia E, Ehrlich M, Massol R, Boucrot E, Brunner C, Kirchhausen T (2006)
Dev Cell 10:839– 850.
34. Seer y JP, Watt FM (2000) Curr Biol 10:1447–1450.
35. Gong Y, Mo C, Fraser SE (2004) Nature 430:689 693.
36. Lechler T, Fuchs E (2005) Nature 437:275–280.
37. Gibson MC, Patel A B, Nagpal R, Perrimon N (2006) Nature 442:1038 –1041.
38. Bastiaens P, Caudron M, Niethammer P, Karsenti E (2006) Trends Cell Biol
16:125–134.
39. Kiger AA, Baum B, Jones S, Jones MR, Coulson A, Echeverri C, Perrimon N
(2003) J Biol 2:27.
40. Eggert US, Kiger AA, Richter C, Perlman ZE, Perrimon N, Mitchison TJ, Field
CM (2004) PLoS Biol 2:e379.
41. Echard A, Hickson GR, Foley E, O’Farrell PH (2004) Curr Biol 14:1685–1693.
42. Bjorklund M, Taipale M, Varjosalo M, Saharinen J, Lahdenpera J, Taipale J
(2006) Nature 439:1009–1013.
43. Gromley A, Yeaman C, Rosa J, Redick S, Chen CT, Mirabelle S, Guha M,
Sillibourne J, Doxsey SJ (2005) Cell 123:75–87.
44. Papoulas O, Hays TS, Sisson JC (2005) Nat Cell Biol 7:612– 618.
45. Lecuit T, Wieschaus E (2000) J Cell Biol 150:849 860.
46. Dhonukshe P, Balusk a F, Schlicht M, Hlavacka A, Samaj J, Friml J, Gadella
TW, Jr (2006) Dev Cell 10:137–150.
47. McNeil PL, Kirchhausen T (2005) Nat Rev Mol Cell Biol 6:499–505.
48. Tapper H, Furuya W, Grinstein S (2002) J Immunol 168:5287–5296.
49. Czibener C, Sherer NM, Becker SM, Pypaert M, Hui E, Chapman ER, Mothes
W, Andrews NW (2006) J Cell Biol 174:997–1007.
50. Braun V, Fraisier V, Raposo G, Hurbain I, Sibarita JB, Chavrier P, Galli T,
Niedergang F (2004) EMBO J 23:4166– 4176.
51. Coupin GT, Muller CD, Remy-Kristensen A, Kuhry JG (1999) J Cell Sci
112(14):2431–2440.
52. Massol RH, Boll W, Griffin AM, K irchhausen T (2006) Proc Natl Acad Sci USA
103:10265–10270.
53. Royle SJ, Bright NA, Lagnado L (2005) Nature 434:1152–1157.
7944
www.pnas.orgcgidoi10.1073pnas.0702511104 Boucrot and Kirchhausen
... In contrast to the well-known function of CME in neuronal differentiation, we know much less about the direct role of CME components in NPC proliferation. In fact, most of the research to date has been conducted in nonneuronal cells, where the activity of endocytic processes was initially suggested to be inhibited during mitosis (Pypaert et al, 1991;Warren, 1993;Raucher & Sheetz, 1999), although later studies have challenged this view (Boucrot & Kirchhausen, 2007;Tacheva-Grigorova et al, 2013). In nonneuronal cells, CME components control mitotic progression independently of their canonical roles in endocytosis and localize to the centrosome, mitotic spindle or midbody to regulate the spindle morphology and/or cytokinesis (Thompson et al, 2004;Royle et al, 2005;Lehtonen et al, 2008;Booth et al, 2011;Olszewski et al, 2014). ...
... Numerous studies have previously knocked down AP-2 (mostly using RNAi-mediated approaches) to investigate its effect on CME (Motley et al, 2003;Diril et al, 2006;Keyel et al, 2008;Lau & Chou, 2008;Boucrot et al, 2010). In these studies, no apparent increase in apoptosis and/or cell division defects were reported, although cells depleted of AP-2µ have been shown to display aberrant spindles (Boucrot & Kirchhausen, 2007). However, most of the studies were performed in cancer or immortalized cell lines with disrupted cell cycle checkpoints. ...
Article
Full-text available
Centrosomes are organelles that nucleate microtubules via the activity of gamma–tubulin ring complexes (γ-TuRC). In the developing brain, centrosome integrity is central to the progression of the neural progenitor cell cycle, and its loss leads to microcephaly. We show that NPCs maintain centrosome integrity via the endocytic adaptor protein complex-2 (AP-2). NPCs lacking AP-2 exhibit defects in centrosome formation and mitotic progression, accompanied by DNA damage and accumulation of p53. This function of AP-2 in regulating the proliferative capacity of NPCs is independent of its role in clathrin-mediated endocytosis and is coupled to its association with the GCP2, GCP3, and GCP4 components of γ-TuRC. We find that AP-2 maintains γ-TuRC organization and regulates centrosome function at the level of MT nucleation. Taken together, our data reveal a novel, noncanonical function of AP-2 in regulating the proliferative capacity of NPCs and open new avenues for the identification of novel therapeutic strategies for the treatment of neurodevelopmental and neurodegenerative disorders with AP-2 complex dysfunction.
... Boucrout (2015) found that the depletion of A1, A2 and A3 endophilin isoforms affected clathrin-independent, dynamin-dependent cellular uptake of G protein-coupled receptors like α2a-and β1-adrenergic receptors, dopaminergic D3 and D4 receptors, and muscarinic acetylcholine receptor 4; the receptor tyrosine kinases EGFR, hepatocyte growth factor receptor, vascular endothelial growth factor receptor, platelet-derived growth factor receptor, nerve growth factor receptor (also known as TNFRSF16) and insulin-like growth factor 1 receptor; as well as the CIE cargo IL-2 receptor . Endophilins A1-A3 can interact with some of these cargo proteins, as previously shown for β1-adrenergic receptor (Boucrot and Kirchhausen, 2007). Since endophilins can induce membrane curvature (Gallop et al., 2006), they may play key roles in the early construction of endocytic pits in these CIE events. ...
... The small GTPase Rab5, phosphatidylinositol 3-kinase (PI3K) and its product phosphatidylinositol-3-phosphate (PtdIns3P) mark the early endosome and are required for its function. The mildly acidic environment in the lumen of early endosomes facilitates conformational changes on proteins and leads to the release of ligands from receptors (Boucrot and Kirchhausen, 2007). ...
Thesis
A host of endocytic pathways exist at the surface of eukaryotic cells, which lead to the internalization of the bulk of membranes along with membrane proteins, signaling receptors, growth factors, and other cargoes (Smith et al. 2017). For decades, the clathrin-mediated pathway has been the major well characterized endocytic process where clathrin polymerizes along with the associated adaptor proteins to include ligand-bound receptors, leading to membrane bending, membrane scission, and endocytosis (Smith et al. 2017). Recently, multiple alternative mechanisms have been uncovered which facilitate the endocytic uptake of cargo molecules and membrane receptors even in the absence of clathrin machinery (Mayor et al.2014). A model of endocytosis that doesn’t require clathrin but rather sugar-binding galectins and glycolipids has been proposed by my host laboratory (Lakshminarayan et al. 2014). Galectins constitute a family of beta-galactoside–binding lectins, which to date consists of 15 members in mammals. Galectins are broadly distributed in a variety of cells and tissues (Leffler et al. 2004). They are translocated from the cytosol to the extracellular space by a process of non-classical secretion (Hughes 1999). Glycosphingolipids (GSLs) are ubiquitous membrane constituents that are subdivided in neutral or acidic fractions. The term GSLs applies to compounds that contain at least one monosaccharide and a ceramide. Of note, the enzyme UDP-glucose ceramide glucosyltransferase (Ugcg) catalyzes the initial step for the biosynthesis of glycosylceramide-based GSLs.Our current working model, which involves glycolipids and lectins, was termed the GL-Lect hypothesis (Johannes et al. 2016). It is backed up by experimental data as described in Ref. (Lakshminarayan et al. 2014) and can be described as follows:i) Monomeric Gal3 binds to glycoproteinsii) Gal3 then starts to oligomerizeiii) Oligomerized Gal3 has the capacity to bind to glycosphingolipids and this may induce clustering of GSLsiv) Gal3-GSL cluster are inducing the invagination of the plasma membrane to generate tubular endocytic pits from which clathrin-independent carriers (CLICs, which are pre-early endosomes) are generated.Oligomeric Gal3 is indeed able to bind to GSLs and to induce membrane deformation (Lakshminarayan et al. 2014) in a similar way the pathogenic lectin Shiga toxin-B subunit (STxB) does. Therefore, both processes could be summarized under the same hypothesis, the GL-Lect hypothesis, where GL stands for the glycosphingolipids (Gb3 for STxB and gangliosides for Gal3) and Lect summarizes the lectins (STxB, Gal3 and possibly others as well).Understanding if this clathrin-independent but Gal3-dependent internalization mechanism is conserved not only in vitro model systems but in vivo is a main challenge in the field of trafficking.We characterized for the first time that in the gut a new mechanism facilitates endocytic uptake of cargo. This mechanism is driven by Galectin3 and operates in intestinal enterocytes for transcytosis like process and is glycosphingolipid dependent. Indeed, we have found that the lactotransferrin (LTF), a Gal3 cargo that we have identified by Mass spec, strongly required Gal3 and GSLs for its efficient endocytosis and its transcytosis like distribution pattern, respectively. Based on these findings in mouse intestinal epithelium, we established a functional in vivo model system where the newly proposed endocytic mechanism termed in our lab as GL-Lect, was physiologically investigated.
... Interestingly, it has also been shown that CME is not completely arrested during natural mitosis but instead on chemically arresting mitosis. 148 This is explained by showing the reduced availability of transferrin receptors on the rounded cell surface keeping CME machinery active at it basal level 149,150 until it stalls at metaphase 151 and regains its activity from telophase. 152 Meanshiwle, the highest plaque density 153 observed to have formed during mitosis in turn aids in respreading of daughter cells after cytokinesis. ...
Article
Full-text available
Cells inhabit a mechanical microenvironment that they continuously sense and adapt to. The plasma membrane (PM), serving as the boundary of the cell, plays a pivotal role in this process of adaptation. In this Review, we begin by examining well-studied processes where mechanoregulation proves significant. Specifically, we highlight examples from the immune system and stem cells, besides discussing processes involving fibroblasts and other cell types. Subsequently, we discuss the common molecular players that facilitate the sensing of the mechanical signal and transform it into a chemical response covering integrins YAP/TAZ and Piezo. We then review how this understanding of molecular elements is leveraged in drug discovery and tissue engineering alongside a discussion of the methodologies used to measure mechanical properties. Focusing on the processes of endocytosis, we discuss how cells may respond to altered membrane mechanics using endo- and exocytosis. Through the process of depleting/adding the membrane area, these could also impact membrane mechanics. We compare pathways from studies illustrating the involvement of endocytosis in mechanoregulation, including clathrin-mediated endocytosis (CME) and the CLIC/GEEC (CG) pathway as central examples. Lastly, we review studies on cell–cell fusion during myogenesis, the mechanical integrity of muscle fibers, and the reported and anticipated roles of various molecular players and processes like endocytosis, thereby emphasizing the significance of mechanoregulation at the PM.
... In animal cells, furrow ingression is directed by a combination of cytoskeletal force generation and membrane trafficking events (reviewed by Pollard, 2010;Neto et al., 2011;Schiel and Prekeris, 2013). Depending on the final dimensions of the daughter cells, there is often a requirement for membrane growth during this division processin addition, this membrane addition may be polarized to direct essential activities required for cytokinesis and abscission (Knutton et al., 1975;Erickson and Trinkaus, 1976;Shuster and Burgess, 2002;Danilchik et al., 2003;Kouranti et al., 2006;Boucrot and Kirchhausen, 2007;Dambournet et al., 2011;Giansanti et al., 2015;Figard et al., 2016). In the cleavage cycles that are present in many animal embryos, this need to rapidly supply membrane is especially apparent and is required to support the fast expansion and remodeling of furrow behaviors. ...
Article
Full-text available
Ingression of the plasma membrane is an essential part of the cell topology-distorting repertoire and a key element in animal cell cytokinesis. Many embryos have rapid cleavage stages in which they are furrowing powerhouses, quickly forming and disassembling cleavage furrows on timescales of just minutes. Previous work has shown that cytoskeletal proteins and membrane trafficking coordinate to drive furrow ingression, but where these membrane stores are derived from and how they are directed to furrowing processes has been less clear. Here, we identify an extensive Rab35/Rab4>Rab39/Klp98A>trans-Golgi network (TGN) endocytic recycling pathway necessary for fast furrow ingression in the Drosophila embryo. Rab39 is present in vesiculotubular compartments at the TGN where it receives endocytically derived cargo through a Rab35/Rab4-dependent pathway. A Kinesin-3 family member, Klp98A, drives the movements and tubulation activities of Rab39, and disruption of this Rab39-Klp98A-Rab35 pathway causes deep furrow ingression defects and genomic instability. These data suggest that an endocytic recycling pathway rapidly remobilizes membrane cargo from the cell surface and directs it to the trans-Golgi network to permit the initiation of new cycles of cleavage furrow formation.
... Cholesterol dependent localization to cleavage furrow PI(4,5,)P2 Regulate membrane to cytoskeleton interaction [196,197] F-BAR domain/Cdc15 Cell middle/contractile ring Anionic PLs Interacts with formin Cdc12 and promote contractile ring formation [65] Gin4 (Nim1 protein kinase) PIPs Septin assembly [198] Golgi phosphoprotein 3 (GOLPH3) ...
Article
Full-text available
The plasma membrane of eukaryotic cells is composed of a large number of lipid species that are laterally segregated into functional domains as well as asymmetrically distributed between the outer and inner leaflets. Additionally, the spatial distribution and organization of these lipids dramatically change in response to various cellular states, such as cell division, differentiation, and apoptosis. Division of one cell into two daughter cells is one of the most fundamental requirements for the sustenance of growth in all living organisms. The successful completion of cytokinesis, the final stage of cell division, is critically dependent on the spatial distribution and organization of specific lipids. In this review, we discuss the properties of various lipid species associated with cytokinesis and the mechanisms involved in their polarization, including forward trafficking, endocytic recycling, local synthesis, and cortical flow models. The differences in lipid species requirements and distribution in mitotic vs. male meiotic cells will be discussed. We will concentrate on sphingolipids and phosphatidylinositols because their transbilayer organization and movement may be linked via the cytoskeleton and thus critically regulate various steps of cytokinesis.
Article
Full-text available
The importance of Echinoderms in Egypt grows day after day, as well as, the usage of the available untapped resources, in particular our interest in mariculture. Sea cucumber fishing was prohibited in Egypt and listed as endangered species, due to overfishing. Although the above decision was right at its time, it is not acceptable to us, due to the economic, nutritional, and pharmaceutical importance of the sea cucumber. So; this work was designed to study the animal well microscopically, apply the artificial induction of autotomy on the animal for the numerical increase and reproduction asexually, and chemically investigate the medical importance of the coelomic fluid of the sea cucumber “Holothuria arenicola”. Materials and methods: the body wall and the coelomic epithelium were studied microscopically. For the animal numerical increase, the autotomy was induced in 3 groups by manual ligation of the body from ¼, ⅓, and ½ of the whole body length of the adults; respectively, in addition to the control group. Side by side with the abovementioned studies, the acellular chemical content of the coelomic fluid was analyzed using GC/MS, and the resulting constituents were furtherly-analyzed by protein docking to investigate the ability of the coelomic fluid to accelerate wound healing. Conclusion: Histologically; the body wall of the sea cucumber’s “Holothuria arenicola” is composed of five layers. Ultrastructurally; the coelomic epithelium small (progenitor) cells (coelomocytes) seem to be hypothetically differentiated into two main types: the neighboring/surrounding cells (that might supply the neighbouring dividing cells with the membranes or any cytoplasmic organelles, hypothetically via exocytosis or endocytic recycling trafficking events or by plasma membrane remodeling), and defence cells that resemble the human neutrophils. Experimentally, the artificially-induced autotomy of the half-body-length ligated group is the only successful group that divides completely. Chemically, we found the presence of bioactive GSK3-β protein in the sea cucumber-extracted coelomic fluid which makes them a valuable resource for wound healing treatment especially in diabetics. Finally, the sea cucumber “Holothuria arenicola” is considered a gift for Egypt, due to its proven significant potential for economic and medical applications, both criteria of significance are concomitant with the sustainable developmental goals and Vision of Egypt 2030.
Thesis
Elevated levels of low-density lipoprotein (LDL) and its derivative lipoprotein(a) (Lp(a)) in the bloodstream are directly implicated in the development and progression of atherosclerotic cardiovascular diseases (ASCVD). LDL homeostasis is mainly regulated by the LDL receptor (LDLR), which is expressed predominantly on the cell surface of hepatocytes where it binds and removes free LDL particles from circulation. Endocytosed LDLR may then either be recycled back to the cell surface or diverted to lysosomes for its degradation. LDLR recycling is therefore critical for efficient LDL uptake. Endocytic recycling is a complex process involving several trafficking steps and many potential molecular mediators. Here I characterized the role of a small GTPase, RAB10, in the trafficking of LDLR. I found that RAB10 depletion inhibits LDLR recycling from endocytic recycling compartment (ERC), thereby reducing surface LDLR abundance and cellular LDL uptake. Further investigation showed that RAB10 also promotes recycling of another cell surface receptor, the transferrin receptor (TFR), but from a different endocytic compartment. Our findings suggest that RAB10 impacts recycling of LDLR and TFR by promoting vesicle trafficking from different intracellular compartments. Although Lp(a) is another casual risk factor for ASCVD, there are no specific Lp(a)-lowering treatments, and their development is limited by a gap in knowledge regarding Lp(a) regulation. Unlike the well-known receptor mediated pathway for LDL homeostasis, the mechanism(s) for cellular uptake of Lp(a) are poorly understood, and several receptors, including LDLR, have been individually studied with conflicting data. To address this gap, we applied a high-throughput whole-genome CRISPR screen to interrogate the regulators of Lp(a) endocytosis in HuH7 cells. Surprisingly, LDLR and other known regulators of LDLR, including SCAP, MBTPS2, MYLIP were among the top hits in the screen. No other receptors, including those previously proposed to serve as Lp(a) receptors, exhibited a functional influence on Lp(a) uptake in this screen. Our results suggest that the LDL receptor is the primary mediator of Lp(a) uptake in hepatocytes. Collectively, the work done in this thesis increases our understanding of receptor mediated trafficking of LDL and Lp(a).
Article
Full-text available
During mitosis, chromosome missegregation and cytokinesis defects have been recognized as hallmarks of cancer cells. Cytoskeletal elements composing the spindle and the contractile ring and their associated proteins play crucial roles in the faithful progression of mitotic cell division. The hypothesis that PGRMC1, most likely as a part of a yet-to-be-defined complex, is involved in the regulation of spindle function and, more broadly, the cytoskeletal machinery driving cell division is particularly appealing. Nevertheless, more than ten years after the preliminary observation that PGRMC1 changes its localization dynamically during meiotic and mitotic cell division, this field of research has remained a niche and needs to be fully explored. To encourage research in this fascinating field, in this review, we will recap the current knowledge on PGRMC1 function during mitotic and meiotic cell division, critically highlighting the strengths and limitations of the experimental approaches used so far. We will focus on known interacting partners as well as new putative associated proteins that have recently arisen in the literature and that might support current as well as new hypotheses of a role for PGRMC1 in specific spindle subcompartments, such as the centrosome, kinetochores, and the midzone/midbody.
Article
Full-text available
Dynamin is one of the major proteins involved in endocytosis. First identified 50 years ago in a genetic screen in Drosophila melanogaster, it has become a central player in many forms of endocytosis, such as clathrin-mediated endocytosis or synaptic vesicle endocytosis, as well as other important cellular processes such as actin remodelling. Decades of work using biochemical and structural studies, cell-free assays, live cell imaging, acute inhibition and genetic studies have led to important insights on its mode of action. Dynamin is a remarkable mechano-GTPase which can do a lot to membranes on its own but which is, in cells, at the centre of a vast protein and lipid network and cannot work in isolation. These results have been synthetized in several important reviews and viewpoints [1–3]. In this review, I will summarize the main features of dynamin structure and function and its central role in membrane remodelling events, and give an update on the latest results.
Article
Full-text available
Indirect immunofluorescence, immunoelectron microscopy, and digestion by protease were used to study intracellular transport of the G protein of vesicular stomatitis virus in mitotic and interphase cells. Quantitation showed that the appearance of G protein on the surface of mitotic cells was inhibited at least 10-fold when compared with that on interphase cells, even though similar amounts of viral protein were being synthesized. This dramatic inhibition, taken together with the simultaneous inhibition of endocytosis (Berlin, R. D., and J. M. Oliver, 1980, J. Cell Biol. 85: 660-671), points to a general cessation of membrane traffic in the mitotic cell.
Article
Full-text available
Synchronized populations of Chinese hamster ovary (CHO) cells in confluent culture have been examined by scanning electron microscopy and their surface changes noted as the cells progress through the cycle. During G1 it is characteristic for cells to show large numbers of microvilli, blebs, and ruffles. Except for the ruffles, these tend to diminish in prominence during S and the cells become relatively smooth as they spread thinly over the substrate. During G2 microvilli increase in number and the cells thicken in anticipation of rounding up for mitosis. It appears that the changes observed here reflect the changing capacity of CHO cells during the cycle to respond to contact with other cells in the population, because, as noted in the succeeding paper (Rubin and Everhart), CHO cells in sparse nonconfluent cultures do not show the same wide range of changes during the cell cycle. Normal, nontransformed cells of equivalent type in confluent culture are essentially devoid of microvilli, blebs, and ruffles. The relation of these surface configurations to the internal structure of the cell is discussed.
Article
Full-text available
The surface morphology of synchronized P815Y mastocytoma cells has been examined by scanning electron microscopy. Early G1 cells are comparatively smooth or light villated, whereas at later stages the surface becomes progressively more villated. In G1 cell most microvilli have a uniform diameter, whereas in S and G2 cells, many microvilli show branching and often originate from much larger surface protuberances. Small "blebs" are seen on the surface of many cells but these structures do not appear to be a characteristic feature of cells at any one stage of the cycle. The presence of microvilli increases the total surface of the cell to such an extent that the ratio of volume to surface area remains constant throughout the cell cycle. The mechanism of cytokinesis is thus a physical one, involving the unfolding of previously accumulated microvilli.
Article
Full-text available
Osmium impregnation was used to determine the number of Golgi apparatus in both interphase and mitotic HeLa cells. The number was found to increase substantially during mitosis to the point where random partitioning alone would explain the nearly equal numbers found in each daughter cell.
Article
Full-text available
Synchronized populations of Chinese hamster ovary (CHO) cells in confluent culture have been examined by scanning electron microscopy and their surface changes noted as the cells progress through the cycle. During G(1) it is characteristic for cells to show large numbers of microvilli, blebs, and ruffles. Except for the ruffles, these tend to diminish in prominence during S and the cells become relatively smooth as they spread thinly over the substrate. During G(2) microvilli increase in number and the cells thicken in anticipation of rounding up for mitosis. It appears that the changes observed here reflect the changing capacity of CHO cells during the cycle to respond to contact with other cells in the population, because, as noted in the succeeding paper (Rubin and Everhart), CHO cells in sparse nonconfluent cultures do not show the same wide range of changes during the cell cycle. Normal, nontransformed cells of equivalent type in confluent culture are essentially devoid of microvilli, blebs, and ruffles. The relation of these surface configurations to the internal structure of the cell is discussed.
Article
HeLa cells arrested in prometaphase were pulse-labeled with [35S]methionine and chased in the absence of nocodazole to allow passage through mitosis and into G1. Transport of histocompatibility antigen (HLA) molecules to the medial- and trans-Golgi cisternae was measured by monitoring the resistance to endoglycosidase H and the acquisition of sialic acid residues, respectively. Transport to the plasma membrane was measured using neuraminidase to remove sialic acid residues on surface HLA molecules. The half-time for transport to each of these compartments was about 65-min longer in cells progressing out of mitosis than in G1 cells. This delay was only 5-min longer than the half-time for the fall in histone H1 kinase activity suggesting that inactivation of the mitotic kinase triggers the resumption of protein transport. The half-time for reassembly of the Golgi stack, measured using stereological procedures, was also 65 min, suggesting that both transport and reassembly are triggered at the same time. However, since reassembly was complete within 5 min, whereas HLA took 25 min to reach the medial-cisterna, we can conclude that the Golgi stack has reassembled by the time HLA reaches it.
Article
Phagocytosis, pinocytosis and the surface distribution of concanavalin A (ConA) have been analyzed during mitosis in several mammalian cell lines. Use of the bisbenzimidazole dye, Hoechst 33258, for chromosome staining after gentle fixation made possible the rapid identification and correlation of mitotic phase with surface properties. Phagocytosis of both opsonized and nonopsonized particles is markedly depressed in mitotic cells of the mouse macrophage cell line J774.1. The uptake of opsonized particles (IgG-coated erythrocytes) is impaired from early prophase through early G1, whereas phagocytosis of nonopsonized particles (latex beads) is restored by telophase. Fluid pinocytosis, determined by the uptake of soluble horseradish peroxidase, is also inhibited during mitosis. Thus peroxidase-containing cytoplasmic vesicles were virtually absent from mid-prophase through telophase in both J774 and Chinese hamster ovary (CHO) cells. Adsorptive pinocytosis of ConA was determined from the different distributions of fluorescence in single cells incubated at 37 degrees C with rhodamine-conjugated ConA (surface and cytoplasmic label), then fixed and further incubated with fluorescein-conjugated anti-ConA (surface only). The separate fluorescence of Hoechst, fluorescein and rhodamine could be optically isolated. In interphase J774 cells, ConA is rapidly internalized into cytoplasmic vesicles. In contrast, ConA is restricted to the plasma membrane from mid-prophase through telophase. In CHO, the depressed pattern of internalization is not fully established until metaphse. The surface distribution of ConA also varied dramatically as a function of mitotic phase. Between mid-prophase and early anaphase, the pattern of surface ConA-receptor complexes is diffuse. Once the cleavage furrow begins to develop, however, ConA moves into the region of the furrow. This was shown in J774, CHO and 3T3 mouse embryonic fibroblasts, and is probably universal. ConA movement into the membrane that overlies the microfilaments of the contractile ring is analogous to similar movements that occur in interphase cells during ConA cap formation and during the development of phagocytic pseudopods. The analogy emphasizes the common functional consequences of microfilament-membrane organization. It is evident that membrane processes which depend upon endocytosis--for example, certain hormone-induced signals--may be interrupted during mitosis. Inhibition of endocytosis thus may be a significant element in the control of cellular activities during mitosis and a strong influence on the properties of the emergent post-mitotic cell.
Article
The increase in surface area that occurs as cells spread from the rounded to the flattened state has been examined in synchronized BHK 21 cells in the scanning electron microscope. Rounded cells, whether in mitosis or dissociated and freshly seeded in culture, are covered with a mixture of folds, blebs, and microvilli. As cells spread, these protuberances disappear, first in the flattening marginal region and progressively submarginally until the entire cell surface is virtually smooth. The estimated surface area of rounded post-mitotic daughter cells, taking microvilli into account, is close to that of fully spread cells 4 h after mitosis. Likewise, rounded early mitotic mother cells, which are also covered with microvilli, have approximately the same surface as fully spread cells just prior to mitosis. These findings suggest that cells possess a membrane reserve in their microvilli and other protuberances which can be utilized for spreading and initiating cell locomotion.
Article
Quantitative ultrastructural and biochemical methods have allowed us to obtain a coherent set of data on the internalization efficiency of the transferrin receptor (TfR). In confluent cell cultures we find that (1) the initial internalization rate of transferrin is approximately 10% per minute, and (2) around 10% of cell-surface TfRs are present in coated pits. From these data a lifetime of coated pits of ca. 1 min is derived. Furthermore, we show that coated pits constitute 1.1-1.4% of the plasma membrane area in confluent cell cultures. Thus, the TfR is concentrated six- to ninefold in coated pits compared to resident plasma membrane proteins. Moreover, we show that the concentration of TfRs in coated pits is cell density dependent, since only around 5% of the receptors are present in coated pits in low-density cultures. Correspondingly, the internalization of TfRs in high-density cell cultures is roughly twice as efficient as that in low-density cell cultures. The reduced TfR internalization efficiency at low cell density is accounted for by a concomitant decrease to 0.55% in the relative surface area occupied by coated pits.
Article
Endocytosis is inhibited during mitosis in A431 cells (Warren et al., 1984) but the site of inhibition is unknown. A quantitative method measuring the extent of budding was used to compare coated pits in interphase and mitotic cells. Every stage of budding found in interphase cells was also found in cells at every stage of mitosis. Flatter coated pits appeared more frequent in mitotic cells but this can be partly, if not entirely, explained by their greater size. We conclude that, if budding is inhibited, inhibition must occur at all stages of the budding process.