You are on page 1of 14

REVIEW ARTICLE

Cytoskeleton, October 2013 70:590–603 (doi: 10.1002/cm.21130)


C
V 2013 Wiley Periodicals, Inc.

How Filopodia Pull: What We Know About the


Mechanics and Dynamics of Filopodia
Thomas Bornschl€
ogl*
Institut Curie, Laboratoire, Physico-Chimie UMR CNRS 168, 11 Rue Pierre et Marie Curie, 75005, Paris, France
Received 16 May 2013; Revised 31 July 2013; Accepted 1 August 2013
Monitoring Editor: Laura Machesky

In recent years, the dynamic, hair-like cell protrusions external mechanical and chemical cues [Mattila and
called filopodia have attracted considerable attention. Lappalainen, 2008; Mellor, 2010]. The most extensively
They have been found in a multitude of different cell studied filopodia are those protruding from the neuronal
types and are often called “sensory organelles,” since growth cone (See Fig. 1A for a schematic drawing). These
they seem to sense the mechanical and chemical envi- were initially discovered in the late 19th century by Ramon
ronment of a cell. Once formed, filopodia can exhibit y Cajal in fixed nervous tissue and were later observed in
complex behavior, they can grow and retract, push or vitro by Ross Harrison, who used the first tissue culture of
pull, and transform into distinct structures. They are dynamic neuron explants [Cajal, 1890; Harrison, 1910;
often found to make first adhesive contact with the Keshishian, 2004; Meldolesi, 2011]. Today, there is a grow-
extracellular matrix, pathogens or with adjacent cells, ing body of evidence that growth cone filopodia are
and to subsequently exert pulling forces. Much is involved in the sensing of chemotactic signals leading to the
known about the cytoskeletal players involved in filopo- directed, path-finding growth of an axon [Gallo and
dia formation, but only recently have we started to Letourneau, 2004; Geraldo and Gordon-Weeks, 2009;
explore the mechanics of filopodia together with the Koleske, 2003]. For instance, it was shown that growth
related cytoskeletal dynamics. This review summarizes cones turning toward a chemoattractant, exhibit an
current advancements in our understanding of the increased number of filopodia on the side that is facing the
mechanics and dynamics of filopodia, with a focus on attractant [Gundersen and Barrett, 1980; Zheng et al.,
the molecular mechanisms behind filopodial force 1996]. Additionally, Cytochalasin-treated growth cones
exertion. V 2013 Wiley Periodicals, Inc.
C with deprived filopodia migrate in a disoriented manner
[Bentley and Toroian-Raymond, 1986]. The physical con-
Key Words: filopodia; actin; retrograde flow; force; mem- tact of a single filopodium with a high affinity substrate,
brane tension
such as a guidepost neuron or a bead coated with nerve
growth factor is sufficient to change the path of a growth
cone [O’Connor et al., 1990; Gallo et al., 1997]. Their
Introduction—The Biological Role of ability to sense guidance cues [Davenport et al., 1993;
Filopodia Shafer et al., 2011], coupled with their ability to apply trac-
tion forces on the substrate via a frictional slippage mecha-
F ilopodia have been observed in a multitude of different
cell types, both in vivo and by using cell cultures on 2D
and 3D substrates in vitro. Under these experimental condi-
nism [Chan and Odde, 2008], would allow the filopodium
to act as a steering mechanism for neuronal growth. Many
of our insights into growth cone guidance are gained from
tions, filopodia are often the first component of the cell to in vitro cell culture experiments, but nonmammalian model
establish contact with extracellular cues, and they are able systems or the extraction of mammalian cortical slices allow
to probe a large area surrounding the cell due to their verification of these findings in vivo [Kalil et al., 2011].
dynamics and hair-like shape. This makes filopodia perfect In the nervous system, filopodia can be involved in other
“cell antennae” and indeed in many instances the primary specialized tasks aside from growth cone guidance. For
biological role of filopodia seems to be the sensing of example, dendritic filopodia have recently been shown to
play a role in the sensation of touch in specialized sensory
*Address Correspondence to: Thomas Bornschl€ogl, Institut Curie, neurons of Drosophila larvae [Tsubouchi et al., 2012]. Strik-
Laboratoire Physico-Chimie, Centre de Recherche, 26 Rue
d’Ulm, Paris F-75248, France. E-mail: bornschl@ph.tum.de
ingly, filopodia can also mature into different structures,
Published online 3 September 2013 in Wiley Online Library they are the precursors of presynaptic boutons at the initial
(wileyonlinelibrary.com). stage of synaptogenesis and they can become spines or

䊏 590
A similar situation arises during dorsal or ventral closure
in developing embryos of different organisms. In vivo con-
focal microscopy showed filopodia in Drosophila embryos
during dorsal closure, where they were implicated in the
zipping of the two opposing leading edge cells [Jacinto
et al., 2000] (See Fig. 1B for a schematic drawing). These
filopodia control the accurate alignment of the opposing
epithelial sheets by creating mutually exclusive contacts
between matching partner cells [Millard and Martin,
2008]. Filopodia have also been observed in C. elegans dur-
ing ventral closure and in migrating muscle cells [Williams-
Masson et al., 1997; Raich et al., 1999; Viveiros et al.,
2011], as well as during sea urchin gastrulation [Miller
et al., 1995]. In all these situations, filopodia seem to fulfill
a sensory and adhesive function toward specific binding
partners. This function, together with the build up of
mechanical force and cell internal signaling, could promote
the proper positioning of the cells within the developing
embryo [Hutson et al., 2003; Lecuit et al., 2011].
Filopodia have been shown to sense signaling molecules a
large distance away from the cell body. Filopodia-like struc-
Fig. 1. Filopodia and forces in biological systems. A) Sketch tures, implicated in cell–cell communication in inverte-
of a migrating growth cone on a 2D substrate. The growth cone
is turning toward the right, e.g., due to the gradient of a chemo- brates, were first observed in the Drosophila wing imaginal
attractant. The filopodia exert traction forces (F) on the substrate disc and were named cytonemes [Ramirez-Weber and
and show asymmetric distribution before turning; more filopodia Kornberg, 1999; Gradilla and Guerrero, 2013]. Cytonemes
are extending toward the chemoattractant. B) Drawing of a Dro- can orient toward the morphogen [Hsiung et al., 2005] and
sophila embryo during the stage of dorsal closure. Filopodia span are specialized for the sensing of specific signaling pathways
the gap between the two opposing epithelial sheets at the position
of zipping (see black square and bottom picture for zoom). Zip- [Roy et al., 2011]. Highly dynamic filopodia have recently
ping forces, together with selective adhesion to matching partner been shown to be critical for tissue patterning in Drosophila.
cells, are important for proper alignment. C) Sketch of well- Here, the physical contact of filopodia can transmit Delta-
adhered cells cultured on a 2D substrate. Filopodia-like cell–cell Notch signaling over long distances, which explains how
bridges between the infected upper cell and a noninfected cell the spacing of bristles on the Drosophila notum is con-
can transport virus particles (red circle). Bacteria, such as Shigella,
can contact the tips of filopodia and induce their retraction in
trolled [De Joussineau et al., 2003; Cohen et al., 2010].
order to get pulled toward the cell for invasion (red ellipse). In a Recently, a similar long-range sensing mechanism of the
similar way, macrophages use filopodia to detect and pull patho- morphogen sonic hedgehog has been observed in the verte-
gens toward the cell body, where phagocytosis occurs. brate limb bud of chick embryos [Sanders et al., 2013].
Similar filopodia-like cell–cell bridges have been impli-
dendrites [Heiman and Shaham, 2010; Hotulainen and cated in the transport of various cargoes ranging from Ca21
Hoogenraad, 2010; Menna et al., 2011]. to entire organelles and are often called tunneling nano-
Besides their importance within the nervous system, filo- tubes in this context [Rustom et al., 2004; Abounit and
podia also play a role in cell–cell adhesion of epithelial tis- Zurzolo, 2012; Sherer, 2013]. Pathogens can hijack filopo-
sues, for example during morphogenesis or wound healing. dia and filopodia-like cell–cell bridges (See Fig. 1C for a
In the situation of wounded epithelia, the implicated cells schematic drawing). For example, viruses have been
often produce an actomyosin cable surrounding the orifice, observed surfing on filopodia-like cell bridges from an
which then closes. Depending on the organism and cell infected cell toward a noninfected cell, thus explaining why
type, filopodial and lamellipodial protrusions can be viral infection is more efficient when cells have physical
observed in the leading edge cells [Garcia-Fernandez et al., contact [Lehmann et al., 2005; Sherer and Mothes, 2008].
2009]. Filopodia seem to be implicated in making first con- Invasive bacteria can hijack filopodia of epithelial cells to
tacts with the opposing cell fronts, where they might serve approach the host cell before infection occurs [Romero
as precursors for the formation of mature cell–cell junctions et al., 2011]. Similarly, macrophages have been shown to
[Vasioukhin et al., 2000; Brevier et al., 2008]. In wounded use filopodia as precursors for phagocytosis. Their filopodia
epithelia, filopodia can also act as long-range sensors for can capture latex beads [Koerten et al., 1980], as well as
cytokines, such as epidermal growth factor [Lidke et al., beads covered with bacterial surface proteins [Kress et al.,
2005]. This cytokine is upregulated at the site of the wound 2007; Zidovska and Sackmann, 2011], and pull them
and leads to enhanced cell migration to support healing. toward the host cell, where phagocytosis can occur.

CYTOSKELETON Bornschl€ogl 591 䊏


Fig. 2. Organization of the actin cytoskeleton in different cells. A) Electron microscopy (EM) of a growth cone from a differenti-
ated B35 rat neuroblastoma cell [Korobova and Svitkina, 2008]. The image shows the typical, fan-like organization with filopodial
actin bundles that are deeply embedded in the dendritic lamellipodial actin network. B) EM of the leading edge of the lamellipodium
of a B16F1 mouse melanoma cell. The protruded filopodium is rooted at its base within the lamellipodial actin network. C) EM of
the same cell type showing two joining actin bundles forming a filopodial k-precursor. The bundled filaments splay apart at the filo-
podial base and are again intimately interwoven with the dendritic lamellipodial network. From Svitkina et al. [2003].

The different filopodia listed above fulfill highly special- 1993], electron microscopy allows to resolve the cytos-
ized tasks. However, they share similarities in their structure keletal organization of filopodia. Figure 2A illustrates the
and they are often built up by the same basic components typical, fan-like cytoskeletal organization of a neuronal
[Faix et al., 2009]. Thus, they likely use a limited set of growth cone with protruding filopodia [Bridgman and Dai-
molecular core components together with an additional, ley, 1989; Schaefer et al., 2002; Korobova and Svitkina,
specific molecular machinery that allows them to fulfill 2008]. Electron microscopy has shown that the filopodial
their sensory tasks [Gupton and Gertler, 2007; Abounit core is made up of 15–30 tightly packed actin filaments,
and Zurzolo, 2012; Gradilla and Guerrero, 2013]. A com- which can exhibit a dense protein complex at the tip [Small
mon function of growth cone, macrophage, and epithelial and Celis, 1978; Lewis and Bridgman, 1992]. In a 2D envi-
filopodia could lie in the sensing of specific binding part- ronment, filopodial filaments are observed in different cell
ners with a subsequent adhesion and build up of retraction types to be deeply rooted in the sheet-like lamellipodia
force. While we have a rather good understanding of the from which they can protrude [Pollard and Borisy, 2003;
basic cytoskeletal players that comprise filopodia, we have Small and Resch, 2005]. The actin filaments at the filopo-
only recently started to explore how these structures exert dial root can be intimately interwoven with the adjacent
forces. In this review, I will summarize experiments address- lamellipodial meshwork (See Figs. 2B and 2C for the struc-
ing these questions. tural organization of the filopodial=lamellipodial interface
in B16-F1 mouse melanoma cells) [Svitkina et al., 2003].
The Structure of Filopodia In the lamellipodium, the branched actin network is organ-
ized in a polarized fashion, with actin filaments pointing
Since their first observation with light microscopy more with their fast-growing barbed end toward the leading edge
than a 100 years ago [Cajal, 1890; Meldolesi, 2011], [Small et al., 1978]. The polarity of an actin filament is
improvements in microscopy techniques have allowed usually identified via the addition of molecular motor
researchers to gain insight into the molecular organization heads, showing a polar decoration of the filament. Recently,
of filopodia. A huge step forward was the invention of elec- this polarity has been observed directly via image analysis of
tron microscopy, together with cell fixation techniques that electron tomographs of Swiss 3T3 cell lamellipodia [Narita
allowed “finger-like processes” with a diameter of only et al., 2012].
200 nm and length of several lm at the leading edge of Filopodia can also protrude from cellular locations other
migrating cells on 2D substrates to be resolved [Porter then the lamellipodium. For example, they can be found at
et al., 1945]. Although atomic force microscopy can be the apical or dorsal cell surface of cells cultured on 2D sub-
used to visualize filopodia of living cells [Parpura et al., strates [Bohil et al., 2006; Planchon et al., 2011] and they

䊏 592 How Filopodia Pull CYTOSKELETON


can protrude from the cellular actin cortex as it can be seen in uous filaments, as shown in Fig. 2B. One exception from
situ [Fraccaroli et al., 2012]. The actin cortex is a 100-nm these conventional filopodia is the Dictyostelium filopo-
thin, isotropic and contractile actin network that is situated dium, which was recently visualized with intact membrane
directly behind the plasma membrane [Salbreux et al., 2012]. by cryo-electron microscopy [Medalia et al., 2007]. In these
The constituent actin filaments are mostly aligned parallel to cells, the actin filaments within the shaft were mostly
the membrane, but a few short perpendicular filaments have aligned parallel to the filopodial axis but showed only an
also been observed [Morone et al., 2006]. In 2D cultures, filo- average length of 200 nm, much shorter than the observed
podia can still protrude from cells with depleted lamellipodia 1-lm long filopodia. Additionally, short actin filaments
[Steffen et al., 2006; Gomez et al., 2007; Korobova and Svit- were observed at the base of these filopodia. These filaments
kina, 2008; Nicholson-Dykstra and Higgs, 2008; Sarmiento are linked to each other at various angles and are not deeply
et al., 2008]. In this scenario, filopodia can form at the edges rooted in the cortical actin network [Medalia et al., 2007].
of concave actin filament bundles that are arranged parallel to Another exception is the dendritic filopodium, the precur-
the plasma membrane [Steffen et al., 2006], which might sor of dendritic spines [Hotulainen and Hoogenraad, 2010]
resemble the geometry of cortically anchored filopodia. How- which has recently been shown to contain a small fraction
ever, most structural studies on filopodia are done with cells of actin filaments pointing with their slow-growing end
on 2D substrates, where they are rooted within the lamellipo- toward the filopodial tip [Korobova and Svitkina, 2010].
dium, and thus little is known about the 3 dimensional ultra- Additionally, immunostaining showed the unusual presence
structure of the cell cortex and how filopodia are anchored of the actin-branching Arp2=3 complex and of myosin II in
within [Ben-Harush et al., 2010]. the filopodial shaft, while the usually abundant cross-linker
Filopodia can form from the polarized lamellipodial actin fascin was absent.
network by two proposed mechanisms, referred to as the The structure of filopodia might not only depend on the
“convergent elongation model” and the “de-novo nuclea- specific cell type, but it might also change in correlation
tion model” [Faix et al., 2009; Yang and Svitkina, 2011]. In with the dynamic state of the filopodium. For example, in
the first model, the branched filaments in the lamellipo- Dictyostelium, growing filopodia appeared straight and
dium, that are formed via Arp2=3 complex-mediated nucle- firmly filled with actin, while retracting filopodia appeared
ation, make k-shaped precursors at the leading edge [Small often bent and contained an almost disassembled actin core
et al., 1998; Svitkina et al., 2003; Yang and Svitkina, 2011] [Medalia et al., 2007]. A recent study, using live cell imag-
and become crosslinked by fascin [Vignjevic et al., 2006] ing correlated with electron tomography on B16-F1 mouse
(See Fig. 2C for an k-shaped precursor). In the “de-novo melanoma cells, showed that retracting filopodia contained
nucleation model,” a parallel actin bundle is newly formed a disintegrated actin bundle in correlation with the presence
and further polymerized at the leading edge of the lamelli- of the actin-depolymerizing factor cofilin [Breitsprecher
podium via the action of e.g., formins [Faix and Rottner, et al., 2011]. In contrast, growing filopodia of the same
2006; Faix et al., 2009; Mellor, 2010]. In both models, the cells showed the conventional, tightly packed and parallel
filopodia formed must contain a bundle of polarized actin actin bundle with no cofilin present.
filaments, pointing with their fast-growing ends toward the
tip of the filopodium, as observed experimentally [Lewis The Dynamics of Filopodia
and Bridgman, 1992]. Within the filopodial shaft, actin fil-
aments can be resolved without disruption throughout the Different proteins regulate filopodial formation, mainte-
filopodium (See Fig 2) [Lewis and Bridgman, 1992]. It is nance and dynamics: proteins that positively control actin
important to note that the fixation techniques used for elec- polymerization, such as formins [Peng et al., 2003; Romero
tron microscopy might lead to slight alterations of the et al., 2004; Pellegrin and Mellor, 2005; Schirenbeck et al.,
observed actin structures [Small et al., 2008]. For example, 2005; Mellor, 2010] and proteins of the Ena=VASP family
if the plasma membrane is removed with Triton before fixa- [Applewhite et al., 2007; Drees and Gertler, 2008] can
tion and negative staining, filopodial actin filaments can localize at filopodia tips. Proteins such as gelsolin [Lu et al.,
show gaps in between themselves and appear as wave-like 1997], capping protein [Mejillano et al., 2004], and EPS8
structures [Lewis and Bridgman, 1992]. In addition, parts [Disanza et al., 2006; Menna et al., 2009] can negatively
of the filopodial actin core are membrane-anchored, so control filopodia formation due to their ability to cap actin
removing the membrane will likely disturb the related [Menna et al., 2011]. Furthermore, membrane-bound pro-
structural features. Cryo-electron tomography circumvents teins from the I-BAR family, which sense and possibly
this problem by rapidly arresting the cellular structure in its induce curved membranes, are implicated in filopodial for-
functional state, thus allowing insights into the actin- mation [Govind et al. 2001; Krugmann et al., 2001; Yama-
membrane interface on a molecular level [Ben-Harush gishi et al., 2004; Millard et al., 2005; Ahmed et al., 2010;
et al., 2010]. Zhao et al., 2011]. Myosin X, a molecular motor, trans-
Filopodia from most cell types are structurally very simi- ports cargo toward the filopodial tip [Berg et al., 2000;
lar, showing a crosslinked actin bundle of polarized, contin- Zhang et al., 2004] and plays a role in filopodial formation

CYTOSKELETON Bornschl€ogl 593 䊏


fied in different cells types (See Fig. 3 movement I, Fig. 4A
and Table I).
The movement of a filopodium that is embedded in the
lamellipodium depends on lamellipodial dynamics. The
cytoskeletal dynamics of lamellipodia are well established in
a wide variety of cell types and show a continuous rearward
flow of the underlying actin network [Pollard and Borisy,
2003; Small and Resch, 2005]. Different proteins control
this retrograde flow. It is driven by the pushing action of
treadmilling actin filaments that polymerize against the
membrane at the leading edge and by contractile forces e.g.,
via myosin II at the lamellipodial base, where actin fila-
ments are depolymerized (See Fig. 4A) [Iwasa and Mullins,
2007; Lai et al., 2008; Bernstein and Bamburg, 2010;
Bugyi and Carlier, 2010].
Retrograde flow was also observed in filopodia. For
example, it can be observed indirectly, e.g., by analyzing the
rearward movement of material on growth cone filopodia
Fig. 3. The dynamics of filopodia. Reported dynamics of filo-
[Sheetz et al., 1992]. The first direct observations of actin
podia that are embedded in the rearward-moving lamellipodium
(thick grey arrow). The most commonly observed movement is filaments moving rearward within growth cone filopodia
linear growth or retraction (I.). Growth or retraction is deter- were made using photo-bleaching and photo-activation
mined by the difference between actin polymerization speed at microscopy on labeled actin [Okabe and Hirokawa, 1991;
the tip and retrograde flow in the filopodium. Sideward move- Mallavarapu and Mitchison, 1999]. Quantification of these
ment of the filopodium (II.) can be explained via orthogonal dynamics showed that filopodial growth and retraction are
rearward flow in the lamellipodium, if the filopodial filaments
are tilted with respect to the cell edge. Filopodia can kink determined by the difference between actin polymerization
within the actin shaft (III) or within the lamellipodium (IV).
Filopodia can also show scanning motion, turning around their
cell anchor point via angular rotations.

[Berg and Cheney, 2002; Watanabe et al., 2010], while


other nonconventional myosins might control membrane-
cytoskeleton adhesion [Nambiar et al., 2010].
Once formed, filopodia are dynamic structures that can
exhibit different modes of movement (See Fig. 3). For
example, they can show angular rotations around their cell
anchor point, which allows the filopodium to “scan” a large
area of the cell environment (Fig. 3, movement V)
[Albrecht-Buehler, 1976; Tamada et al., 2010; Zidovska
and Sackmann, 2011]. Filopodia that fold back onto the
cell in such a motion have been shown to become part of Fig. 4. Filopodial force exertion. A) Sketch of a linearly grow-
lateral, contractile actin bundles within the lamellipodium ing or retracting filopodium as shown in Fig. 3(I). In the lamel-
lipodium, the polymerization of actin at the leading edge (red
of migrating fish fibroblasts and B16 melanoma cells [Nem- stripe) induces pushing forces against the cell membrane. This
ethova et al., 2008]. They can also move laterally with action, together with the contraction and depolymerization of
respect to the lamellipodial cell front (Fig. 3, movement II), actin (e.g., via myosin II and cofilin) at the lamellipodial base
which can be explained by their tilted insertion within the (green area), leads to a continuous retrograde flow of the den-
rearward-flowing lamellipodial actin network [Oldenbourg dritic actin network. Since the filopodial and lamellipodial
structures are connected, friction could drive the retrograde flow
et al., 2000]. The filopodial root, when embedded in the in the filopodium. Filopodia can then exert pulling forces using
lamellipodium, can kink [Nemethova et al., 2008] (Fig. 3, the dynamics of the rearward-flowing actin (Fa), together with
movement IV), and a similar “jack-knife” mechanism inward forces arising from membrane tension (Fm). Filopodial
within the shaft of filopodia was observed in macrophages tips can exert pushing forces via actin polymerization at the tip.
and growth cone filopodia when plated on 2D substrates B) Different geometries of filopodial adhesion. Reported pulling
forces are high (nN) if the filopodium exhibits a large adhesive
(Fig. 3, movement III) [Sheetz et al. 1992; Kress et al., contact zone along the entire shaft. Reported pulling forces were
2007]. However, the most common motions of filopodia 1003 smaller for geometries with decreased adhesive contacts,
are linear growth and retraction, which have been quanti- such as contact only at the tip (from left to right).

䊏 594 How Filopodia Pull CYTOSKELETON


Table I. Retrograde Flow in Filopodia
Growth Retraction Retrograde
Filopodia=cell type Species (nm=s) (nm=s) flow (nm=s) Citation
Growth cone Aplysia, Chick, 17–170 17–170 10–100 Reviewed in Geraldo
filopodia Rat, Direct (photo-bleaching/ and Gordon-Weeks
and Mouse photo-activation of [2009]
labeled actin)
Embryonic cerebral Mouse 240 210 260 [Sheetz et al., 1992]
cortex filopodia Indirect (movement
of material)
Dendritic hippocampal Rat 0–17 0–17 20 [Tatavarty et al., 2012]
E18 filopodia Direct (PALM)
Cervix adenocarcinoma Human 135 84 (bacteria 22 [Romero et al., 2011]
(HeLa) filopodia induced) Direct
(photo-bleaching)
HeLa 100 10 15, 23 [Berg and Cheney,
Indirect (myosin 2002; Kerber et al.,
X movement) 2009]
HeLa 42 [Schelhaas et al., 2008]
Direct (photo-activation
and indirect virus
movement)
HeLa 5–50 [ur Rehman et al., 2012]
Indirect
(syndecan surfing)
Hela, epidermoid carcinoma Human 26, 18, 27 [Lidke et al., 2005]
cells (A431) Indirect (movement
and breast carcinoma of epidermal growth
cells (MCF7) factor coated
quantum dots)
Epidermal keratinocyte Human 40 13 20 vs. 42 [Schafer et al., 2010;
filopodia Direct (photo-bleaching Schafer et al., 2011]
of adhesive vs.
nonadhesive filopodia)
Kidney cell COS7 African green 70 10 30 [Watanabe et al., 2010]
filopodia monkey Indirect (myosin X
movement)
Cytonemes between African green 12 [Sherer et al., 2007]
kidney cells monkey Indirect
Cos-1 and XC and rat (virus surfing)
sarcoma cells
Embryonic kidney cell Human 30, 17 [Lehmann et al., 2005]
(HEK 293) Indirect
filopodia (virus surfing)
Quantifications of retrograde flow within filopodia of different cell types. Retrograde flow within filopodia of neuronal growth cones is well stud-
ied and is summarized in Geraldo and Gordon-Weeks [2009]. In addition, retrograde flow in filopodia of HeLa cells is now also well quantified
and was directly observed via photo-bleaching and photo-activation experiments. The measurement of e.g., rearward movement of myosin X or
viruses along filopodia can indicate the presence of retrograde actin flow indirectly and are also listed. Retrograde flow was also observed but not
quantified in filopodial shafts of B16F1 mouse melanoma cells [Applewhite et al., 2007] and filopodial filaments embedded in the B16F1 lamelli-
podia move backwards with approximately 30 nm=s [Vignjevic et al., 2006]. The table also summarizes observed average speeds of linearly grow-
ing or retracting filopodia.

rate at the tip and the speed of retrograde flow (Fig. 4A) switching from protrusion to retraction, correlated with a
[Mallavarapu and Mitchison, 1999]. In these experiments, change of the actin polymerization rate at the tip, while the
the majority of the changes in filopodial dynamics, such as retrograde flow speed stayed constant. This suggests that

CYTOSKELETON Bornschl€ogl 595 䊏


the tip-polymerization rate is the main parameter control- polymerizing actin filament is able to produce pushing
ling filopodial dynamics [Mallavarapu and Mitchison, forces of 1 pN [Footer et al., 2007] and slightly higher
1999]. Since then, photo-bleaching or photo-activation of forces of 1.3 pN if its polymerization is assisted by formin
filopodial actin filaments has become a common tool to [Kovar and Pollard, 2004; Berro et al., 2007], which is
quantify retrograde flow, which has led to a good under- often found at filopodial tips. A crosslinked actin bundle
standing of cytoskeletal dynamics within growth cone filo- can resist buckling and therefore produce enough force for
podia [Geraldo and Gordon-Weeks, 2009]. Retrograde filopodial growth via actin polymerization against the mem-
flow was also observed in filopodia of other cell lines either brane at the tip. However, dependent on the number of fila-
indirectly (e.g., via surfing of virus particles [Lehmann ments in the actin core, buckling and the diffusion of G-
et al., 2005]) or directly e.g., via photo-bleaching (See actin toward the tip can be limiting factors for filopodia
Table I). In most cases reported so far, the speed of filopo- outgrowth [Mogilner and Rubinstein, 2005; Atilgan et al.,
dial retraction is slower than the internal retrograde flow 2006; Lan and Papoian, 2008]. By placing optically
speed of actin. These results implicate retrograde flow, trapped, nonadhesive beads in front of filopodia that are
together with reduced actin polymerization speed at the tip, protruding from growth cones of rat dorsal root ganglia,
as the major mechanism for filopodial retraction, although pushing forces of 1–2 pN have been observed [Cojoc et al.,
additional depolymerization of actin from the tip might 2007]. This indicates that the internal actin core is able to
also occur [Breitsprecher et al., 2011]. While retrograde counterbalance membrane tension and to exert additional
flow seems to be a common feature of filopodia, its pres- small pushing forces on external objects.
ence might depend on the particular cell line or the state of It is important to note that actin may not be the only fac-
development of the filopodium. For example, it was shown tor that can push out membrane tubes. Membrane binding
that the retrograde flow observed in dendritic filopodia van- proteins containing an I-BAR domain could be able to
ishes after maturation of the filopodium into a dendritic directly deform the plasma membrane [Suetsugu et al.
spine [Tatavarty et al., 2012]. 2006; Mattila et al., 2007; Ahmed et al., 2010; Zhao et al.,
2011]. For example, overexpression of IRSp53, a member
The Mechanics of Filopodia of the family of I-BAR domain proteins, was shown to
induce actin-free membrane tubes at the tips of filopodia
In their natural environment, filopodia can make first con- from B16F1 melanoma cells [Yang et al., 2009]. Although
tact with other cells, the extracellular matrix or pathogens. these tubes might be mechanically unstable without subse-
The adhesion to a substrate together with its underlying quent support via the actin core, highly concentrated I-
actin dynamics allows the filopodia to exert pushing or pull- BAR proteins could act as precursors for filopodial growth
ing forces. Surprisingly, relatively little is known about the via active deformation of the membrane.
strength with which filopodia of different cells can pull and
how they exert forces. Recent experiments give first insights
into the working mechanisms of the filopodial pulling and Pulling Back With Force
pushing machinery. In most physiological situations where filopodia have estab-
lished adhesive contact to extracellular substrates, as for exam-
Pushing out the Membrane ple during cell migration or wound healing, filopodia seem to
The mechanics of filopodia are defined by the distinct pull (See Fig. 1). Researchers have only recently started to
mechanical properties of its two basic constituents: the actin quantify the pulling forces exerted by filopodia. A first esti-
bundle comprising the filopodial core and the surrounding mate of filopodial pulling forces was obtained by observing
plasma membrane. In order to increase its length, the filo- chick sensory growth cones that attached with their filopodial
podial actin core has to grow inside the plasma membrane tips to perpendicular neurites and subsequently pulled on
tube that has to be pushed out. The plasma membrane is them [Heidemann et al., 1990]. In a prior publication, the
constantly under tension [Gauthier et al., 2012; Diz- same group measured the forces that are needed to deflect
Munoz et al., 2013], thus pushing out a membrane tube neurites using calibrated glass needles [Dennerll et al., 1989],
requires force. The force needed to extrude empty plasma thus allowing them to estimate filopodial pulling forces to be
membrane tubes from the cell e.g., with an optical trap are between 500 and 900 pN [Heidemann et al., 1990].
in the range of 5–30 pN for different cell lines [Dai and One potential means of quantifying forces exerted by
Sheetz, 1995; Sheetz, 2001]. Since the diameter of filopodia cells adhering to a substrate is traction-force microscopy.
is similar to the diameter of empty membrane tubes, cells For this technique, cells are plated on soft substrates with
likely have to push against 10 pN in order to extend a well-calibrated elastic moduli containing marker beads that
filopodium [Derenyi et al., 2002; Daniels and Turner, allow recording of local deformations [Dembo and Wang,
2005; Nambiar et al., 2010]. This protrusion force can be 1999; Schwarz and Gardel, 2012]. From the marker dis-
produced, for example, by the polymerization of actin at placements, local stress in the substrate can be calculated.
the filopodial tip. In vitro experiments showed that a single Using this technique, it was shown that filopodia from

䊏 596 How Filopodia Pull CYTOSKELETON


cervical ganglion neurons from mice can exert maximal [Daniels and Turner, 2005]. Although the diameter of
pulling traction forces in the order of 1 nN on the substrate retracting filopodia has been observed to increase [Heide-
[Bridgman et al., 2001]. Traction-force microscopy was also mann et al., 1990], it would still be too small to entirely
used to show that retracting Dictyostelium filopodia exert account for forces in the nN range.
pulling forces via their distal tip portion [Iwadate and Therefore, it must primarily be the cytoskeleton that pro-
Yumura, 2008]. The observed stresses were in the range of duces the reported high retraction forces. Filopodial actin
0.3 kPa, which leads to an estimated pulling force of 100 filaments could couple to the lamellipodial actin network
pN, if we assume that the filopodium used a contact area of and use active rearward forces produced by lamellipodial
1.0 3 0.2 lm2 to pull on the substrate. retrograde flow (Fig. 4A). One argument that points toward
Elastic, hair-like nanowires can also be used to estimate high frictional coupling is the observation that filopodial
pulling forces of filopodia. Human dermal foreskin fibro- actin filaments flow backward together with the adjacent
blasts were plated on substrates that exhibited islands of lamellipodial network in growth cones, chick embryo fibro-
“nanowire bushels.” Subsequently, electron microscopy was blasts, and B16F1 cells [Mallavarapu and Mitchison, 1999;
used to show that filopodia attaching to nanowires are able Medeiros et al., 2006; Vignjevic et al., 2006; Anderson
to bend these structures. By measuring the deflection of et al., 2008]. Frictional coupling would also explain the
nanowires, it was estimated that filopodia can pull with contribution of myosin II to filopodial pulling. In migrat-
forces of up to 2 nN [Albuschies and Vogel, 2013]. ing cells, myosin II is usually found in the lamella at the
Filopodia from macrophages are known to bind with rear of the lamellipodium (see green area in Fig. 4A) [Ponti
their tips to bacteria in order to pull them toward the cell et al., 2004; Vicente-Manzanares et al., 2009; Wilson et al.,
body, where phagocytosis occurs. By replacing the bacteria 2010], where it occasionally interacts with the roots of
with an optically trapped bead, it was shown that these filo- deeply embedded filopodia [Anderson et al., 2008; Neme-
podia pull in a step-wise manner and pulling drastically thova et al., 2008; Xue et al., 2010]. Similarly, in growth
slowed down at relatively small forces of 15 pN [Kress cones, myosin II localizes at the rear of the lamellipodium
et al., 2007]. Additionally, similar measurements using at the interface between P- and C-domains [Rochlin et al.,
magnetic tweezers showed that macrophage filopodia stop 1995; Medeiros et al., 2006]. Myosin II is a major driving
pulling and elongate with a force-independent, visco-elastic force behind retrograde flow [Cai et al., 2006; Vicente-
behavior, if external forces between 20 and 600 pN are Manzanares et al., 2009], but it only accounts for half of
applied [Vonna et al., 2007; Zidovska and Sackmann, the retrograde flow in growth cones, with the other half
2011]. Similarly, filopodia from HeLa cells have been being produced by the pushing action of actin filaments
shown to pull with their tips on optically trapped beads and against the membrane at the lamellipodial leading edge
this pulling can be stalled at small forces around 15 pN [Craig et al., 2012; Medeiros et al., 2006]. The contribu-
[Romero et al., 2012]. tion of myosin II to lamellipodial retrograde flow would
Interestingly, the pulling forces of filopodia listed above explain why filopodia from growth cones of myosin IIB
exhibit a high degree of variability: the strongest reported knock-out mice pulled less strongly on the substrate [Bridg-
pulling forces in the nN range are similar to those exerted man et al., 2001].
by focal adhesions, highly specialized structures that trans- The observation that the speed of retrograde flow in the
mit forces to the extracellular substrate [Shemesh et al., filopodium decreases when they exert higher forces on the
2009; Wolfenson et al., 2009; Schwarz and Gardel, 2012]. substrate agrees with the model of frictional coupling. For
In contrast, in other experiments filopodial pulling was example, growth cone filopodia that are attached to soft
slowed down, inverted or stalled by forces of only 15 pN, substrates show reduced speed of retrograde flow as com-
a force that could be produced by only a few molecular pared to stiff substrates [Chan and Odde, 2008]. This
motors. One remaining question is where these differences observation was modeled with a stochastic “motor-clutch”
come from. In the following, I will summarize first experi- system that transduces filopodial traction forces with differ-
ments that can give hints to the answer. ent efficiency to the substrate in dependence on the sub-
strates’ stiffness. On soft substrates, filopodia pulled via a
load-and-fail behavior, transducing high traction forces that
How Filopodia Pull Back correlated with a slow retrograde flow [Chan and Odde,
Filopodial rearward force can arise from membrane tension 2008]. Similarly, in migrating keratinocytes it was shown
that leads to an inward force at the filopodial tip and from that retrograde flow in filopodia slowed down if they con-
active pulling via the retrograde flow of the actin cytoskele- tained premature, small focal complexes adhering to the
ton in the cortex. However, forces on the order of 1–2 nN substrate [Schafer et al., 2010]. Interestingly, these small
are unlikely to arise from membrane tension. Those forces, focal complexes can mature later on into focal adhesions
that are on the order of 10 pN for empty plasma mem- [Nemethova et al., 2008; Schafer et al., 2010] that apply
brane tubes [Sheetz, 2001], increase to a first approxima- traction forces to the substrate that depend on the speed of
tion linearly with respect to the filopodial diameter lamellipodial actin flow [Gardel et al., 2008; Shemesh

CYTOSKELETON Bornschl€ogl 597 䊏


et al., 2009; Wolfenson et al., 2009]. Ultimately, in order be the case for macrophages [Kress et al., 2007; Zidovska
to verify if friction between filopodial and lamellipodial and Sackmann, 2011] and it explains the dynamics of ster-
actin networks is responsible for filopodial force produc- eocilia, a structure that shares some basic similarities with
tion, experiments recording flow speeds in both actin struc- filopodia. These rod-shaped membrane protrusions show
tures under controlled loads would be necessary. retrograde actin flow that is comparable to that observed in
How can one explain the observed differences of maxi- filopodia, but with a 10003 lower speed [Rzadzinska et al.,
mal filopodial pulling forces that range between 20 and 2004]. Friction at the base should therefore be much
2000 pN? One explanation would be different organization smaller as compared to filopodia. A constant length of ster-
of the cytoskeletal actin network adjacent to filopodial eocilia can then be accomplished if frictional forces at the
roots. This organization depends on the cell type and on base counterbalance forces arising from membrane tension
the environment in which the cell is embedded [Baker and [Prost et al., 2007].
Chen, 2012]. For cells spread on a 2D substrate, a large In the future, exact quantification of these parameters,
contact zone between filopodial roots and the lamellipo- together with the identification and quantification of the
dium would allow high friction and thus high pulling molecular players controlling filopodial mechanics and
forces. In contrast, less adhesive cells, cells with depleted dynamics, will help to further decipher how filopodia
lamellipodia [Steffen et al., 2006] or cells embedded in a accomplish their impressive, multifunctional tasks.
3D environment [Baker and Chen, 2012] could show a dif-
ferent actin organization at filopodial roots with less fric- Conclusions and Perspectives
tional coupling and direct interaction of molecular motors.
This could explain why filopodial retraction from less Fluorescence microscopy techniques such as confocal and
adhesive macrophages, slows down against small forces of speckle microscopy [Danuser and Waterman-Storer, 2006]
only 15 pN [Kress et al., 2007]. However, the driving force make it possible to record the dynamics of the filopodial
behind filopodial retraction in macrophages still needs to and lamellipodial actin cytoskeleton, while traction-force
be identified, since inhibition of myosin II and the use of microscopy can be used to probe filopodial mechanics
myosin V and myosin VI knock-outs continued to show [Chan and Odde, 2008; Kraning-Rush et al., 2012]. In the
step-like retraction [Kress et al., 2007]. future, combinations of new high-resolution techniques,
A second explanation for the observed dispersion in filo- together with force spectroscopy [Ahmed, 2011; Brenner
podial pulling forces is the adhesion strength between the et al., 2011] will yield additional insights into the relation-
filopodium and the substrate (See Fig. 4B). The adhesion ship between the cytoskeletal dynamics and the mechanics
strength depends on the type and total amount of the impli- of filopodia. These techniques will shed new light on the
cated cell-adhesion receptors such as integrins [Letourneau mechanisms filopodia employ in the regulation of force, and
and Shattuck, 1989; Steketee and Tosney, 2002; Zhang how these adhesive structures differ from mature, mechano-
et al., 2004]. In the experiments that reported high pulling sensory focal adhesions [Chan and Odde, 2008; Plotnikov
forces, filopodia were able to adhere over a large area. For et al., 2012]. The combination of live cell imaging and elec-
example on 2D substrates used e.g., in traction-force micros- tron tomography will help to further decipher how the
copy, the filopodia can build adhesive contacts over its whole structure of filopodia and protein content depend on their
length (Fig. 4B, left image) [Bridgman et al., 2001; Chan actual dynamic state. A very important task will be to apply
and Odde, 2008; Schafer et al., 2010]. In contrast, in the our knowledge gained on 2D substrates to better understand
experiments reporting small filopodia pulling forces, the function of the cells cytoskeleton in a more physiological
micrometer-sized beads were used as a substrate that only relevant 3D environment [Baker and Chen, 2012].
contacts a small portion of the filopodium at the tip (Fig.
4B, right image) [Kress et al., 2007; Zidovska and Sack- Acknowledgments
mann, 2011; Romero et al., 2012]. In this configuration it I thank D. Vignjevic and P. Bassereau for discussions and for
was shown that the observed, average pulling forces can be careful reading of the manuscript. Thanks to T. Svitkina for
further decreased by decreasing the concentration of providing the EM images. Thanks to M. Simunovic and M.
filopodia-substrate links [Romero et al., 2012]. In the con- Bussonnier for proofreading and for help with Illlustrator.
text of a fibrillar 3D-environment, small dendritic extensions I acknowledge the “Human Frontier Science Program” for
of fibroblasts, neuron and glia cells have been shown to form funding.
an entangled contact [Jiang and Grinnell, 2005; Sorkin
et al., 2009] and a recent publication models how the References
increase of contact zone between filopodia and substrate can Abounit S, Zurzolo C. 2012. Wiring through tunneling nanotubes-
lead to higher traction forces [Albuschies and Vogel, 2013]. from electrical signals to organelle transfer. J Cell Sci 125(Pt 5):
It is important to note, that force-contributions from the 1089–1098.
membrane tension become important if they get compara- Ahmed S. 2011. Nanoscopy of cell architecture: the actin-
ble to actin-based forces produced in the cortex. This might membrane interface. Bioarchitecture 1(1):32–38.

䊏 598 How Filopodia Pull CYTOSKELETON


Ahmed S, Goh WI, Bu W. 2010. I-BAR domains, IRSp53 and filo- muscle myosin IIA-dependent force inhibits cell spreading and
podium formation. Semin Cell Dev Biol 21(4):350–356. drives F-actin flow. Biophys J 91(10):3907–3920.
Albrecht-Buehler G. 1976. Filopodia of spreading 3T3 cells. Do Cajal SR. 1890. Sobre la aparicion de las expanciones celulares en
they have a substrate-exploring function? J Cell Biol 69(2):275– la medulla embrionaria. Gag Sanit Barc (12):413–419.
286.
Chan CE, Odde DJ. 2008. Traction dynamics of filopodia on com-
Albuschies J, Vogel V. 2013. The role of filopodia in the recogni- pliant substrates. Science 322(5908):1687–1691.
tion of nanotopographies. Sci Rep 3:1658.
Cohen M, Georgiou M, Stevenson NL, Miodownik M, Baum B. 2010.
Anderson TW, Vaughan AN, Cramer LP. 2008. Retrograde flow Dynamic filopodia transmit intermittent Delta-Notch signaling to drive
and myosin II activity within the leading cell edge deliver F-actin pattern refinement during lateral inhibition. Dev Cell 19(1):78–89.
to the lamella to seed the formation of graded polarity actomyosin
II filament bundles in migrating fibroblasts. Mol Biol Cell 19(11): Cojoc D, Difato F, Ferrari E, Shahapure RB, Laishram J, Righi M,
5006–5018. Di Fabrizio EM, Torre V. 2007. Properties of the force exerted by
filopodia and lamellipodia and the involvement of cytoskeletal com-
Applewhite DA, Barzik M, Kojima S, Svitkina TM, Gertler FB, ponents. PloS one 2(10):e1072.
Borisy GG. 2007. Ena=VASP proteins have an anti-capping inde-
pendent function in filopodia formation. Mol Biol Cell 18(7): Craig EM, Van Goor D, Forscher P, Mogilner A. 2012. Membrane
2579–2591. tension, myosin force, and actin turnover maintain actin treadmill
in the nerve growth cone. Biophys J 102(7):1503–1513.
Atilgan E, Wirtz D, Sun SX. 2006. Mechanics and dynamics of
actin-driven thin membrane protrusions. Biophys J 90(1):65–76. Dai J, Sheetz MP. 1995. Mechanical properties of neuronal growth
cone membranes studied by tether formation with laser optical
Baker BM, Chen CS. 2012. Deconstructing the third dimension: tweezers. Biophys J 68(3):988–996.
how 3D culture microenvironments alter cellular cues. J Cell Sci
125(Pt 13):3015–3024. Daniels DR, Turner MS. 2005. Spicules and the effect of rigid rods
on enclosing membrane tubes. Phys Rev Lett 95(23):238101.
Ben-Harush K, Maimon T, Patla I, Villa E, Medalia O. 2010. Vis-
ualizing cellular processes at the molecular level by cryo-electron Danuser G, Waterman-Storer CM. 2006. Quantitative fluorescent
tomography. J Cell Sci 123(Pt 1):7–12. speckle microscopy of cytoskeleton dynamics. Annu Rev Biophys
Biomol Struct 35:361–387.
Bentley D, Toroian-Raymond A. 1986. Disoriented pathfinding by
pioneer neurone growth cones deprived of filopodia by cytochalasin Davenport RW, Dou P, Rehder V, Kater SB. 1993. A sensory role
treatment. Nature 323(6090):712–715. for neuronal growth cone filopodia. Nature 361(6414):721–724.
Berg JS, Cheney RE. 2002. Myosin-X is an unconventional myosin De Joussineau C, Soule J, Martin M, Anguille C, Montcourrier P,
that undergoes intrafilopodial motility. Nat Cell Biol 4(3):246–250. Alexandre D. 2003. Delta-promoted filopodia mediate long-range
lateral inhibition in Drosophila. Nature 426(6966):555–559.
Berg JS, Derfler BH, Pennisi CM, Corey DP, Cheney RE. 2000.
Myosin-X, a novel myosin with pleckstrin homology domains, asso- Dembo M, Wang YL. 1999. Stresses at the cell-to-substrate inter-
ciates with regions of dynamic actin. J Cell Sci 113 Pt 19:3439– face during locomotion of fibroblasts. Biophys J 76(4):2307–2316.
3451.
Dennerll TJ, Lamoureux P, Buxbaum RE, Heidemann SR. 1989.
Bernstein BW, Bamburg JR. 2010. ADF=cofilin: a functional node The cytomechanics of axonal elongation and retraction. J Cell Biol
in cell biology. Trends Cell Biol 20(4):187–195. 109(6 Pt 1):3073–3083.
Berro J, Michelot A, Blanchoin L, Kovar DR, Martiel JL. 2007. Derenyi I, Julicher F, Prost J. 2002. Formation and interaction of
Attachment conditions control actin filament buckling and the pro- membrane tubes. Phys Rev Lett 88(23):238101.
duction of forces. Biophys J 92(7):2546–2558.
Disanza A, Mantoani S, Hertzog M, Gerboth S, Frittoli E, Steffen
Bohil AB, Robertson BW, Cheney RE. 2006. Myosin-X is a molec- A, Berhoerster K, Kreienkamp HJ, Milanesi F, Di Fiore PP, et al.
ular motor that functions in filopodia formation. Proc Natl Acad 2006. Regulation of cell shape by Cdc42 is mediated by the syner-
Sci USA 103(33):12411–12416. gic actin-bundling activity of the Eps8-IRSp53 complex. Nat Cell
Breitsprecher D, Koestler SA, Chizhov I, Nemethova M, Mueller J, Biol 8(12):1337–1347.
Goode BL, Small JV, Rottner K, Faix J. 2011. Cofilin cooperates Diz-Munoz A, Fletcher DA, Weiner OD. 2013. Use the force:
with fascin to disassemble filopodial actin filaments. J Cell Sci membrane tension as an organizer of cell shape and motility. Trends
124(Pt 19):3305–3318. Cell Biol 23(2):47–53.
Brenner MD, Zhou R, Ha T. 2011. Forcing a connection: impacts Drees F, Gertler FB. 2008. Ena=VASP: proteins at the tip of the
of single-molecule force spectroscopy on in vivo tension sensing. nervous system. Curr Opin Neurobiol 18(1):53–59.
Biopolymers 95(5):332–344.
Faix J, Rottner K. 2006. The making of filopodia. Curr Opin Cell
Brevier J, Montero D, Svitkina T, Riveline D. 2008. The asymmet- Biol 18(1):18–25.
ric self-assembly mechanism of adherens junctions: a cellular push-
pull unit. Phys Biol 5(1):016005. Faix J, Breitsprecher D, Stradal TE, Rottner K. 2009. Filopodia:
complex models for simple rods. Int J Biochem Cell Biol 41(8-9):
Bridgman PC, Dailey ME. 1989. The organization of myosin and 1656–1664.
actin in rapid frozen nerve growth cones. J Cell Biol 108(1):95–109.
Footer MJ, Kerssemakers JW, Theriot JA, Dogterom M. 2007.
Bridgman PC, Dave S, Asnes CF, Tullio AN, Adelstein RS. 2001. Direct measurement of force generation by actin filament polymer-
Myosin IIB is required for growth cone motility. J Neurosci 21(16): ization using an optical trap. Proc Natl Acad Sci USA 104(7):
6159–6169. 2181–2186.
Bugyi B, Carlier MF. 2010. Control of actin filament treadmilling Fraccaroli A, Franco CA, Rognoni E, Neto F, Rehberg M, Aszodi
in cell motility. Annu Rev Biophys 39:449–470. A, Wedlich-Soldner R, Pohl U, Gerhardt H, Montanez E. 2012.
Cai Y, Biais N, Giannone G, Tanase M, Jiang G, Hofman JM, Visualization of endothelial actin cytoskeleton in the mouse retina.
Wiggins CH, Silberzan P, Buguin A, Ladoux B, et al. 2006. Non- PloS one 7(10):e47488.

CYTOSKELETON Bornschl€ogl 599 䊏


Gallo G, Letourneau PC. 2004. Regulation of growth cone actin Kalil K, Li L, Hutchins BI. 2011. Signaling mechanisms in cortical
filaments by guidance cues. J Neurobiol 58(1):92–102. axon growth, guidance, and branching. Front Neuroanat 5:62.
Gallo G, Lefcort FB, Letourneau PC. 1997. The trkA receptor Kerber ML, Jacobs DT, Campagnola L, Dunn BD, Yin T, Sousa
mediates growth cone turning toward a localized source of nerve AD, Quintero OA, Cheney RE. 2009. A novel form of motility in
growth factor. J Neurosci 17(14):5445–5454. filopodia revealed by imaging myosin-X at the single-molecule level.
Garcia-Fernandez B, Campos I, Geiger J, Santos AC, Jacinto A. Curr Biol 19(11):967–973.
2009. Epithelial resealing. Int J Dev Biol 53(8-10):1549–1556. Keshishian H. 2004. Ross Harrison’s “The outgrowth of the nerve
Gardel ML, Sabass B, Ji L, Danuser G, Schwarz US, Waterman fiber as a mode of protoplasmic movement”. J Exp Zool A Comp
CM. 2008. Traction stress in focal adhesions correlates biphasically Exp Biol 301(3):201–203.
with actin retrograde flow speed. J Cell Biol 183(6):999–1005. Koerten HK, Ploem JS, Daems WT. 1980. Ingestion of latex beads
Gauthier NC, Masters TA, Sheetz MP. 2012. Mechanical feedback by filopodia of adherent mouse peritoneal macrophages. A scanning
between membrane tension and dynamics. Trends Cell Biol 22(10): electron microscopical and reflection contrast microscopical study.
527–535. Exp Cell Res 128(2):470–475.

Geraldo S, Gordon-Weeks PR. 2009. Cytoskeletal dynamics in Koleske AJ. 2003. Do filopodia enable the growth cone to find its
growth-cone steering. J Cell Sci 122(Pt 20):3595–3604. way? Sci STKE 2003(183):pe20.
Gomez TS, Kumar K, Medeiros RB, Shimizu Y, Leibson PJ, Korobova F, Svitkina T. 2008. Arp2=3 complex is important for
Billadeau DD. 2007. Formins regulate the actin-related protein 2=3 filopodia formation, growth cone motility, and neuritogenesis in
complex-independent polarization of the centrosome to the immu- neuronal cells. Mol Biol Cell 19(4):1561–1574.
nological synapse. Immunity 26(2):177–190. Korobova F, Svitkina T. 2010. Molecular architecture of synaptic
Govind S, Kozma R, Monfries C, Lim L, Ahmed S. 2001. actin cytoskeleton in hippocampal neurons reveals a mechanism of
Cdc42Hs facilitates cytoskeletal reorganization and neurite out- dendritic spine morphogenesis. Mol Biol Cell 21(1):165–176.
growth by localizing the 58-kD insulin receptor substrate to fila- Kovar DR, Pollard TD. 2004. Insertional assembly of actin filament
mentous actin. J Cell Biol 152(3):579–594. barbed ends in association with formins produces piconewton
Gradilla AC, Guerrero I. 2013. Cytoneme-mediated cell-to-cell sig- forces. Proc Natl Acad Sci USA 101(41):14725–14730.
naling during development. Cell Tissue Res 352(1):59–66. Kraning-Rush CM, Carey SP, Califano JP, Reinhart-King CA.
Gundersen RW, Barrett JN. 1980. Characterization of the turning 2012. Quantifying traction stresses in adherent cells. Methods Cell
response of dorsal root neurites toward nerve growth factor. J Cell Biol 110:139–178.
Biol 87(3 Pt 1):546–554.
Kress H, Stelzer EH, Holzer D, Buss F, Griffiths G, Rohrbach A.
Gupton SL, Gertler FB. 2007. Filopodia: the fingers that do the 2007. Filopodia act as phagocytic tentacles and pull with discrete
walking. Sci STKE 2007(400):re5. steps and a load-dependent velocity. Proc Natl Acad Sci USA
104(28):11633–11638.
Harrison RG. 1910. The outgrowth of the nerve fiber as a mode of
protoplasmic movement. J Exp Zool 9:787–848. Krugmann S, Jordens I, Gevaert K, Driessens M, Vandekerckhove
Heidemann SR, Lamoureux P, Buxbaum RE. 1990. Growth cone J, Hall A. 2001. Cdc42 induces filopodia by promoting the forma-
behavior and production of traction force. J Cell Biol 111(5 Pt 1): tion of an IRSp53:Mena complex. Curr Biol 11(21):1645–1655.
1949–1957. Lai FP, Szczodrak M, Block J, Faix J, Breitsprecher D, Mannherz
Heiman MG, Shaham S. 2010. Twigs into branches: how a filopo- HG, Stradal TE, Dunn GA, Small JV, Rottner K. 2008. Arp2=3
dium becomes a dendrite. Curr Opin Neurobiol 20(1):86–91. complex interactions and actin network turnover in lamellipodia.
EMBO J 27(7):982–992.
Hotulainen P, Hoogenraad CC. 2010. Actin in dendritic spines:
connecting dynamics to function. J Cell Biol 189(4):619–629. Lan Y, Papoian GA. 2008. The stochastic dynamics of filopodial
growth. Biophys J 94(10):3839–3852.
Hsiung F, Ramirez-Weber FA, Iwaki DD, Kornberg TB. 2005.
Dependence of Drosophila wing imaginal disc cytonemes on Lecuit T, Lenne PF, Munro E. 2011. Force generation, transmis-
Decapentaplegic. Nature 437(7058):560–563. sion, and integration during cell and tissue morphogenesis. Annu
Rev Cell Dev Biol 27:157–184.
Hutson MS, Tokutake Y, Chang MS, Bloor JW, Venakides S,
Kiehart DP, Edwards GS. 2003. Forces for morphogenesis investi- Lehmann MJ, Sherer NM, Marks CB, Pypaert M, Mothes W.
gated with laser microsurgery and quantitative modeling. Science 2005. Actin- and myosin-driven movement of viruses along filopo-
300(5616):145–149. dia precedes their entry into cells. Journal Cell Biol 170(2):317–
325.
Iwadate Y, Yumura S. 2008. Molecular dynamics and forces of a
motile cell simultaneously visualized by TIRF and force microscop- Letourneau PC, Shattuck TA. 1989. Distribution and possible
ies. BioTechniques 44(6):739–750. interactions of actin-associated proteins and cell adhesion molecules
of nerve growth cones. Development 105(3):505–519.
Iwasa JH, Mullins RD. 2007. Spatial and temporal relationships
between actin-filament nucleation, capping, and disassembly. Curr Lewis AK, Bridgman PC. 1992. Nerve growth cone lamellipodia
Biol 17(5):395–406. contain two populations of actin filaments that differ in organiza-
tion and polarity. J Cell Biol 119(5):1219–1243.
Jacinto A, Wood W, Balayo T, Turmaine M, Martinez-Arias A,
Martin P. 2000. Dynamic actin-based epithelial adhesion and cell Lidke DS, Lidke KA, Rieger B, Jovin TM, Arndt-Jovin DJ. 2005.
matching during Drosophila dorsal closure. Curr Biol 10(22):1420– Reaching out for signals: filopodia sense EGF and respond by
1426. directed retrograde transport of activated receptors. J Cell Biol
170(4):619–626.
Jiang H, Grinnell F. 2005. Cell-matrix entanglement and mechani-
cal anchorage of fibroblasts in three-dimensional collagen matrices. Lu M, Witke W, Kwiatkowski DJ, Kosik KS. 1997. Delayed retrac-
Mol Biol Cell 16(11):5070–5076. tion of filopodia in gelsolin null mice. J Cell Biol 138(6):1279–1287.

䊏 600 How Filopodia Pull CYTOSKELETON


Mallavarapu A, Mitchison T. 1999. Regulated actin cytoskeleton Okabe S, Hirokawa N. 1991. Actin dynamics in growth cones. J
assembly at filopodium tips controls their extension and retraction. Neurosci 11(7):1918–1929.
J Cell Biol 146(5):1097–1106. Oldenbourg R, Katoh K, Danuser G. 2000. Mechanism of lateral
Mattila PK, Lappalainen P. 2008. Filopodia: molecular architecture movement of filopodia and radial actin bundles across neuronal
and cellular functions. Nat Rev Mol Cell Biol 9(6):446–454. growth cones. Biophys J 78(3):1176–1182.
Mattila PK, Pykalainen A, Saarikangas J, Paavilainen VO, Vihinen Parpura V, Haydon PG, Henderson E. 1993. Three-dimensional
H, Jokitalo E, Lappalainen P. 2007. Missing-in-metastasis and imaging of living neurons and glia with the atomic force micro-
IRSp53 deform PI(4,5)P2-rich membranes by an inverse BAR scope. J Cell Sci 104 (Pt 2):427–432.
domain-like mechanism. J Cell Biol 176(7):953–964. Pellegrin S, Mellor H. 2005. The Rho family GTPase Rif induces
Medalia O, Beck M, Ecke M, Weber I, Neujahr R, Baumeister W, filopodia through mDia2. Curr Biol 15(2):129–133.
Gerisch G. 2007. Organization of actin networks in intact filopo- Peng J, Wallar BJ, Flanders A, Swiatek PJ, Alberts AS. 2003. Dis-
dia. Curr Biol 17(1):79–84. ruption of the Diaphanous-related formin Drf1 gene encoding
Medeiros NA, Burnette DT, Forscher P. 2006. Myosin II functions mDia1 reveals a role for Drf3 as an effector for Cdc42. Curr Biol
in actin-bundle turnover in neuronal growth cones. Nat Cell Biol 13(7):534–545.
8(3):215–226. Planchon TA, Gao L, Milkie DE, Davidson MW, Galbraith JA,
Mejillano MR, Kojima S, Applewhite DA, Gertler FB, Svitkina Galbraith CG, Betzig E. 2011. Rapid three-dimensional isotropic
TM, Borisy GG. 2004. Lamellipodial versus filopodial mode of the imaging of living cells using Bessel beam plane illumination. Nat
actin nanomachinery: pivotal role of the filament barbed end. Cell Methods 8(5):417–423.
118(3):363–373. Plotnikov SV, Pasapera AM, Sabass B, Waterman CM. 2012. Force
Meldolesi J. 2011. Neurite outgrowth: this process, first discovered fluctuations within focal adhesions mediate ECM-rigidity sensing to
by Santiago Ramon y Cajal, is sustained by the exocytosis of two guide directed cell migration. Cell 151(7):1513–1527.
distinct types of vesicles. Brain Res Rev 66(1-2):246–255. Pollard TD, Borisy GG. 2003. Cellular motility driven by assembly
Mellor H. 2010. The role of formins in filopodia formation. Bio- and disassembly of actin filaments. Cell 112(4):453–465.
chim Biophys Acta 1803(2):191–200. Ponti A, Machacek M, Gupton SL, Waterman-Storer CM, Danuser
Menna E, Disanza A, Cagnoli C, Schenk U, Gelsomino G, Frittoli E, G. 2004. Two distinct actin networks drive the protrusion of
Hertzog M, Offenhauser N, Sawallisch C, Kreienkamp HJ, et al. 2009. migrating cells. Science 305(5691):1782–1786.
Eps8 regulates axonal filopodia in hippocampal neurons in response to Porter KR, Claude A, Fullam EF. 1945. A study of tissue culture
brain-derived neurotrophic factor (BDNF). PLoS Biol 7(6):e1000138. cells by electron microscopy: methods and preliminary observations.
Menna E, Fossati G, Scita G, Matteoli M. 2011. From filopodia to J Exp Med 81(3):233–246.
synapses: the role of actin-capping and anti-capping proteins. Eur J Prost J, Barbetta C, Joanny JF. 2007. Dynamical control of the shape
Neurosci 34(10):1655–1662. and size of stereocilia and microvilli. Biophys J 93(4):1124–1133.
Millard TH, Martin P. 2008. Dynamic analysis of filopodial inter- Raich WB, Agbunag C, Hardin J. 1999. Rapid epithelial-sheet seal-
actions during the zippering phase of Drosophila dorsal closure. ing in the Caenorhabditis elegans embryo requires cadherin-
Development 135(4):621–626. dependent filopodial priming. Curr Biol 9(20):1139–1146.
Millard TH, Bompard G, Heung MY, Dafforn TR, Scott DJ, Ramirez-Weber FA, Kornberg TB. 1999. Cytonemes: cellular proc-
Machesky LM, Futterer K. 2005. Structural basis of filopodia for- esses that project to the principal signaling center in Drosophila
mation induced by the IRSp53=MIM homology domain of human imaginal discs. Cell 97(5):599–607.
IRSp53. EMBO J 24(2):240–250.
Rochlin MW, Itoh K, Adelstein RS, Bridgman PC. 1995. Localiza-
Miller J, Fraser SE, McClay D. 1995. Dynamics of thin filopodia tion of myosin II A and B isoforms in cultured neurons. J Cell Sci
during sea urchin gastrulation. Development 121(8):2501–2511. 108 (Pt 12):3661–3670.
Mogilner A, Rubinstein B. 2005. The physics of filopodial protru- Romero S, Le Clainche C, Didry D, Egile C, Pantaloni D, Carlier
sion. Biophys J 89(2):782–795. MF. 2004. Formin is a processive motor that requires profilin to
Morone N, Fujiwara T, Murase K, Kasai RS, Ike H, Yuasa S, accelerate actin assembly and associated ATP hydrolysis. Cell
Usukura J, Kusumi A. 2006. Three-dimensional reconstruction of 119(3):419–429.
the membrane skeleton at the plasma membrane interface by elec- Romero S, Grompone G, Carayol N, Mounier J, Guadagnini S,
tron tomography. J Cell Biol 174(6):851–862. Prevost MC, Sansonetti PJ, Van Nhieu GT. 2011. ATP-mediated
Nambiar R, McConnell RE, Tyska MJ. 2010. Myosin motor func- Erk1=2 activation stimulates bacterial capture by filopodia, which
tion: the ins and outs of actin-based membrane protrusions. Cell precedes Shigella invasion of epithelial cells. Cell Host Microbe
Mol Life Sci 67(8):1239–1254. 9(6):508–519.
Narita A, Mueller J, Urban E, Vinzenz M, Small JV, Maeda Y. Romero S, Quatela A, Bornschl€ogl T, Guadagnini S, Bassereau P,
2012. Direct determination of actin polarity in the cell. J Mol Biol Tran Van Nhieu G. 2012. Filopodium retraction is controlled by
419(5):359–368. adhesion to its tip. J Cell Sci 125(Pt 21):4999–5004.
Nemethova M, Auinger S, Small JV. 2008. Building the actin cyto- Roy S, Hsiung F, Kornberg TB. 2011. Specificity of Drosophila
skeleton: filopodia contribute to the construction of contractile bun- cytonemes for distinct signaling pathways. Science 332(6027):354–
dles in the lamella. J Cell Biol 180(6):1233–1244. 358.
Nicholson-Dykstra SM, Higgs HN. 2008. Arp2 depletion inhibits Rustom A, Saffrich R, Markovic I, Walther P, Gerdes HH. 2004.
sheet-like protrusions but not linear protrusions of fibroblasts and Nanotubular highways for intercellular organelle transport. Science
lymphocytes. Cell Motil Cytoskeleton 65(11):904–922. 303(5660):1007–1010.
O’Connor TP, Duerr JS, Bentley D. 1990. Pioneer growth cone Rzadzinska AK, Schneider ME, Davies C, Riordan GP, Kachar B.
steering decisions mediated by single filopodial contacts in situ. J 2004. An actin molecular treadmill and myosins maintain stereocilia
Neurosci 10(12):3935–3946. functional architecture and self-renewal. J Cell Biol 164(6):887–897.

CYTOSKELETON Bornschl€ogl 601 䊏


Salbreux G, Charras G, Paluch E. 2012. Actin cortex mechanics Small JV, Isenberg G, Celis JE. 1978. Polarity of actin at the lead-
and cellular morphogenesis. Trends Cell Biol 22(10):536–545. ing edge of cultured cells. Nature 272(5654):638–639.
Sanders TA, Llagostera E, Barna M. 2013. Specialized filopodia Small JV, Rottner K, Kaverina I, Anderson KI. 1998. Assembling
direct long-range transport of SHH during vertebrate tissue pattern- an actin cytoskeleton for cell attachment and movement. Biochim
ing. Nature 497(7451):628–632. Biophys 1404(3):271–281.
Sarmiento C, Wang W, Dovas A, Yamaguchi H, Sidani M, El-Sibai Small JV, Auinger S, Nemethova M, Koestler S, Goldie KN,
M, Desmarais V, Holman HA, Kitchen S, Backer JM, et al. 2008. Hoenger A, Resch GP. 2008. Unravelling the structure of the lamel-
WASP family members and formin proteins coordinate regulation of lipodium. J Microsc 231(3):479–485.
cell protrusions in carcinoma cells. J Cell Biol 180(6):1245–1260. Sorkin R, Greenbaum A, David-Pur M, Anava S, Ayali A, Ben-
Schaefer AW, Kabir N, Forscher P. 2002. Filopodia and actin arcs Jacob E, Hanein Y. 2009. Process entanglement as a neuronal
guide the assembly and transport of two populations of microtu- anchorage mechanism to rough surfaces. Nanotechnol 20(1):
bules with unique dynamic parameters in neuronal growth cones. J 015101.
Cell Biol 158(1):139–152. Steffen A, Faix J, Resch GP, Linkner J, Wehland J, Small JV,
Schafer C, Born S, Mohl C, Houben S, Kirchgessner N, Merkel R, Rottner K, Stradal TE. 2006. Filopodia formation in the absence of
Hoffmann B. 2010. The key feature for early migratory processes: functional WAVE-and Arp2=3-complexes. Mol Biol Cell 17(6):
Dependence of adhesion, actin bundles, force generation and trans- 2581–2591.
mission on filopodia. Cell Adhes Migr 4(2):215–225. Steketee MB, Tosney KW. 2002. Three functionally distinct adhe-
Schafer C, Faust U, Kirchgessner N, Merkel R, Hoffmann B. 2011. sions in filopodia: shaft adhesions control lamellar extension. J Neu-
The filopodium: a stable structure with highly regulated repetitive rosci 22(18):8071–8083.
cycles of elongation and persistence depending on the actin cross- Suetsugu S, Murayama K, Sakamoto A, Hanawa-Suetsugu K, Seto
linker fascin. Cell Adhes Migr 5(5):431–438. A, Oikawa T, Mishima C, Shirouzu M, Takenawa T, Yokoyama S.
Schelhaas M, Ewers H, Rajamaki ML, Day PM, Schiller JT, 2006. The RAC binding domain=IRSp53-MIM homology domain
Helenius A. 2008. Human papillomavirus type 16 entry: retrograde of IRSp53 induces RAC-dependent membrane deformation. J Biol
cell surface transport along actin-rich protrusions. PLoS Pathog Chemy 281(46):35347–35358.
4(9):e1000148. Svitkina TM, Bulanova EA, Chaga OY, Vignjevic DM, Kojima S,
Schirenbeck A, Bretschneider T, Arasada R, Schleicher M, Faix J. Vasiliev JM, Borisy GG. 2003. Mechanism of filopodia initiation by
2005. The Diaphanous-related formin dDia2 is required for the reorganization of a dendritic network. J Cell Biol 160(3):409–421.
formation and maintenance of filopodia. Nat Cell Biol 7(6):619– Tamada A, Kawase S, Murakami F, Kamiguchi H. 2010. Autono-
625. mous right-screw rotation of growth cone filopodia drives neurite
Schwarz US, Gardel ML. 2012. United we stand: integrating the turning. J Cell Biol 188(3):429–441.
actin cytoskeleton and cell-matrix adhesions in cellular mechano- Tatavarty V, Das S, Yu J. 2012. Polarization of actin cytoskeleton is
transduction. J Cell Sci 125(Pt 13):3051–3060. reduced in dendritic protrusions during early spine development in
Shafer B, Onishi K, Lo C, Colakoglu G, Zou Y. 2011. Vangl2 pro- hippocampal neuron. Mol Biol Cell 23(16):3167–3177.
motes Wnt=planar cell polarity-like signaling by antagonizing Dvl1- Tsubouchi A, Caldwell JC, Tracey WD. 2012. Dendritic filopodia,
mediated feedback inhibition in growth cone guidance. Dev Cell Ripped Pocket, NOMPC, and NMDARs contribute to the sense of
20(2):177–191. touch in Drosophila larvae. Curr Biol 22(22):2124–2134.
Sheetz MP. 2001. Cell control by membrane-cytoskeleton adhesion. ur Rehman Z, Sjollema KA, Kuipers J, Hoekstra D, Zuhorn IS.
Nat Rev Mol Cell Biol 2(5):392–396. 2012. Nonviral gene delivery vectors use syndecan-dependent trans-
Sheetz MP, Wayne DB, Pearlman AL. 1992. Extension of filopodia port mechanisms in filopodia to reach the cell surface. ACS Nano
by motor-dependent actin assembly. Cell Motil Cytoskeleton 22(3): 6(8):7521–7532.
160–169. Vasioukhin V, Bauer C, Yin M, Fuchs E. 2000. Directed actin
Shemesh T, Verkhovsky AB, Svitkina TM, Bershadsky AD, Kozlov polymerization is the driving force for epithelial cell-cell adhesion.
MM. 2009. Role of focal adhesions and mechanical stresses in the Cell 100(2):209–019.
formation and progression of the lamellipodium-lamellum interface Vicente-Manzanares M, Ma X, Adelstein RS, Horwitz AR. 2009.
[corrected]. Biophys J 97(5):1254–1264. Non-muscle myosin II takes centre stage in cell adhesion and
Sherer NM. 2013. Long-distance relationships: do membrane nano- migration. Nat Rev Mol Cell Biol 10(11):778–790.
tubes regulate cell-cell communication and disease progression? Mol Vignjevic D, Kojima S, Aratyn Y, Danciu O, Svitkina T, Borisy
Biol Cell 24(8):1095–1098. GG. 2006. Role of fascin in filopodial protrusion. J Cell Biol
Sherer NM, Mothes W. 2008. Cytonemes and tunneling nanotu- 174(6):863–875.
bules in cell-cell communication and viral pathogenesis. Trends Cell Viveiros R, Hutter H, Moerman DG. 2011. Membrane extensions
Biol 18(9):414–420. are associated with proper anterior migration of muscle cells during
Sherer NM, Lehmann MJ, Jimenez-Soto LF, Horensavitz C, Caenorhabditis elegans embryogenesis. Dev Biol 358(1):189–200.
Pypaert M, Mothes W. 2007. Retroviruses can establish filopodial Vonna L, Wiedemann A, Aepfelbacher M, Sackmann E. 2007.
bridges for efficient cell-to-cell transmission. Nat Cell Biol 9(3): Micromechanics of filopodia mediated capture of pathogens by
310–315. macrophages. Eur Biophys J 36(2):145–151.
Small JV, Celis JE. 1978. Filament arrangements in negatively Watanabe TM, Tokuo H, Gonda K, Higuchi H, Ikebe M. 2010.
stained cultured cells: the organization of actin. Cytobiologie 16(2): Myosin-X induces filopodia by multiple elongation mechanism. J
308–325. Biol Chem 285(25):19605–19614.
Small JV, Resch GP. 2005. The comings and goings of actin: cou- Williams-Masson EM, Malik AN, Hardin J. 1997. An actin-
pling protrusion and retraction in cell motility. Curr Opin Cell mediated two-step mechanism is required for ventral enclosure of
Biol 17(5):517–523. the C. elegans hypodermis. Development 124(15):2889–2901.

䊏 602 How Filopodia Pull CYTOSKELETON


Wilson CA, Tsuchida MA, Allen GM, Barnhart EL, Applegate KT, Yang C, Hoelzle M, Disanza A, Scita G, Svitkina T. 2009. Coordi-
Yam PT, Ji L, Keren K, Danuser G, Theriot JA. 2010. Myosin II nation of membrane and actin cytoskeleton dynamics during filopo-
contributes to cell-scale actin network treadmilling through network dia protrusion. PloS One 4(5):e5678.
disassembly. Nature 465(7296):373–377.
Zhang H, Berg JS, Li Z, Wang Y, Lang P, Sousa AD, Bhaskar A,
Wolfenson H, Henis YI, Geiger B, Bershadsky AD. 2009. The heel
Cheney RE, Stromblad S. 2004. Myosin-X provides a motor-based
and toe of the cell’s foot: a multifaceted approach for understanding
link between integrins and the cytoskeleton. Nature Cell Biol 6(6):
the structure and dynamics of focal adhesions. Cell Motil Cytoskel-
523–531.
eton 66(11):1017–1029.
Xue F, Janzen DM, Knecht DA. 2010. Contribution of Filopodia Zhao H, Pykalainen A, Lappalainen P. 2011. I-BAR domain pro-
to Cell Migration: A Mechanical Link between Protrusion and teins: linking actin and plasma membrane dynamics. Curr Opin
Contraction. Int J Cell Biol 2010:507821. Cell Biol 23(1):14–21.
Yamagishi A, Masuda M, Ohki T, Onishi H, Mochizuki N. 2004.
Zheng JQ, Wan JJ, Poo MM. 1996. Essential role of filopodia in
A novel actin bundling=filopodium-forming domain conserved in
chemotropic turning of nerve growth cone induced by a glutamate
insulin receptor tyrosine kinase substrate p53 and missing in metas-
gradient. J Neurosci 16(3):1140–1149.
tasis protein. J Biol Chem 279(15):14929–14936.
Yang C, Svitkina T. 2011. Filopodia initiation: focus on the Zidovska A, Sackmann E. 2011. On the mechanical stabilization of
Arp2=3 complex and formins. Cell Adhes Migr 5(5):402–408. filopodia. Biophys J 100(6):1428–1437.

CYTOSKELETON Bornschl€ogl 603 䊏

You might also like