ArticlePDF Available

Extracellular Matrix- and Cytoskeleton-Dependent Changes in Cell Shape and Stiffness

Authors:

Abstract and Figures

Cell spreading is correlated with changes in important cell functions including DNA synthesis, motility, and differentiation. Spreading is accompanied by a complex reorganization of the cytoskeleton that can be related to changes in cell stiffness. While cytoskeletal organization and the resulting cell stiffness have been studied in motile cells such as fibroblasts, less is known of these events in nonmigratory, epithelial cells. Hence, we examined the relationship between cell function, spreading, and stiffness, as measured by atomic force microscopy. Cell stiffness increased with spreading on a high density of fibronectin (1000 ng/cm(2)) but remained low in cells that stayed rounded on a low fibronectin density (1 ng/cm(2)). Disrupting actin or myosin had the same effect of inhibiting spreading, but had different effects on stiffness. Disrupting f-actin assembly lowered both stiffness and spreading, while inhibiting myosin light chain kinase inhibited spreading but increased cell stiffness. However, disrupting either actin or myosin inhibited DNA synthesis. These results demonstrate the relationship between cell stiffness and spreading in hepatocytes. They specifically show that normal actin and myosin function is required for hepatocyte spreading and DNA synthesis and demonstrate opposing effects on cell stiffness upon disruption of actin and myosin.
Content may be subject to copyright.
Extracellular Matrix- and Cytoskeleton-Dependent Changes
in Cell Shape and Stiffness
Kiran Bhadriraju*,1 and Linda K. Hansen*,,2
*Biomedical Engineering Institute, MMC 609 and Department of Laboratory Medicine and Pathology, MMC 609,
420 Delaware Street S.E., University of Minnesota, Minneapolis, Minnesota 55455
Cell spreading is correlated with changes in impor-
tant cell functions including DNA synthesis, motility,
and differentiation. Spreading is accompanied by a
complex reorganization of the cytoskeleton that can
be related to changes in cell stiffness. While cytoskel-
etal organization and the resulting cell stiffness have
been studied in motile cells such as fibroblasts, less is
known of these events in nonmigratory, epithelial
cells. Hence, we examined the relationship between
cell function, spreading, and stiffness, as measured by
atomic force microscopy. Cell stiffness increased with
spreading on a high density of fibronectin (1000 ng/
cm2) but remained low in cells that stayed rounded on
a low fibronectin density (1 ng/cm2). Disrupting actin
or myosin had the same effect of inhibiting spreading,
but had different effects on stiffness. Disrupting f-ac-
tin assembly lowered both stiffness and spreading,
while inhibiting myosin light chain kinase inhibited
spreading but increased cell stiffness. However, dis-
rupting either actin or myosin inhibited DNA synthe-
sis. These results demonstrate the relationship be-
tween cell stiffness and spreading in hepatocytes.
They specifically show that normal actin and myosin
function is required for hepatocyte spreading and
DNA synthesis and demonstrate opposing effects on
cell stiffness upon disruption of actin and myosin.
© 2002 Elsevier Science (USA)
Key Words: actin; atomic force microscopy; cell
shape; cell spreading; cytoskeleton; extracellular ma-
trix; growth; hepatocyte; myosin; stiffness.
INTRODUCTION
Cell spreading on adhesive proteins such as fibronec-
tin, laminin, or collagen is accompanied by changes in
the structure and composition of the cytoskeleton,
which in turn accommodate and facilitate the change
in shape [1, 2]. Spreading is also accompanied by dis-
tinct changes in cell behavior depending on the cell
type; cells that are allowed to spread undergo growth
and dedifferentiation while cells that are constrained
to remain rounded in culture undergo either apoptosis
or differentiation. Several cell types exhibit this strik-
ing shape–function relationship, including hepatocytes
[3], mouse mammary epithelial cells [4], endothelial
cells [5], fibroblasts [6], and adipocytes [7].
Hepatocytes are the principal cell type of the liver
constituting up to 90% of adult liver by weight, and
they perform several critical metabolic functions in-
cluding the synthesis and secretion of bile and several
serum factors including fibronectin, fibrinogen, trans-
ferrin, and albumin. In vitro, hepatocytes cultured on a
low density of fibronectin, collagen, or laminin [3], or
on a synthetic RGD peptide [8], remain rounded and
exhibit enhanced differentiated function. This rounded
morphology is associated with low DNA synthesis. On
the other hand, when allowed to spread well on a high
density of fibronectin, collagen, or laminin, they dedif-
ferentiate and exhibit enhanced DNA synthesis [3].
Hepatocytes similarly exhibit low DNA synthesis and
enhanced differentiated function when a rounded mor-
phology is induced by disrupting f-actin assembly us-
ing cytochalasin D [9], further demonstrating a connec-
tion between cytoskeletal structure and cell growth.
Disrupting microtubules has a less striking effect on
growth or differentiation [9]. Since the motor protein
myosin associates with actin filaments in nonmuscle
cells [10], it is likely that some of the effects of disrupt-
ing the actin cytoskeleton are in fact due to the disrup-
tion of actomyosin assemblies. Little information, how-
ever, exists on how hepatocyte spreading or function is
influenced upon disrupting myosin activity. When we
measured hepatocyte myosin activity by several meth-
ods, we found a consistent increase with cell spreading
of myosin association with the cytoskeleton, light chain
phosphorylation, and ATPase activity (Bhadriraju and
Hansen, manuscript in preparation). However, hepa-
tocytes do not present prominent stress fibers as seen
in fibroblasts or endothelial cells and are generally not
regarded as force-generating cells, since they possess
neither contractile nor motile behavior. Hence, hepa-
1Current address: Johns Hopkins University School of Medicine,
720 Rutland Avenue, Traylor 715, Baltimore, MD 21205.
2To whom correspondence and reprint requests should be ad-
dressed. Fax: (612) 625-1121. E-mail: hanse066@tc.umn.edu.
920014-4827/02 $35.00
© 2002 Elsevier Science (USA)
All rights reserved.
Experimental Cell Research 278, 92–100 (2002)
doi:10.1006/excr.2002.5557
tocytes presented a unique interest from the perspec-
tive of studying stiffness changes related to shape and
function.
Over the past several years, a high-resolution sur-
face imaging instrument called the atomic force micro-
scope (AFM)3has been increasingly used for measuring
the stiffness of small samples such as living cells [11,
12]. In its essence, the AFM consists of a small exible
cantilever whose deection can be measured with sub-
nanometer accuracy as it interacts with the sample
surface. Knowing the cantilever stiffness, these deec-
tions can be translated into forces on the sample, which
then can be used to measure an apparent sample stiff-
ness assuming a mathematical model for the mechan-
ical behavior of the sample [13]. The AFM has been
used for measuring the stiffness of several different
biological samples including cardiac myocytes [14],
cholinergic synaptic vesicles [15], and gelatin [16].
Techniques that use force mapping have been invalu-
able in generating stiffness maps of biological surfaces
[15, 17, 18]. Using such force mapping techniques, it
has been shown that disrupting the actin cytoskeleton
with cytochalasin D reduces cell stiffness while dis-
rupting the microtubule network does not have a sig-
nicant effect on stiffness [19]. While such studies have
been useful in understanding stiffness distribution
over cells, there are few studies relating global changes
in cell shape to stiffness and function or in response to
changes in myosin activity. The present study was
undertaken with the aim of correlating stiffness mea-
surements with spreading and growth. The correlation
between stiffness and cell spreading was determined.
In addition, the effects of disruption of actin or myosin
activity on both DNA synthesis and cell stiffness were
assessed. These ndings are discussed in relation to
possible underlying mechanistic events.
MATERIALS AND METHODS
Cell culture. Hepatocytes were harvested by collagenase perfu-
sion of adult rat livers [20] and used immediately after harvest. Cells
were cultured in a hormonally dened medium containing saturat-
ing amounts of growth factors and supplements: 10 ng/ml epidermal
growth factor (Collaborative Research, Bedford, MA), 5 nM dexa-
methasone (Sigma, St. Louis, MO), 20 mU/ml insulin (Sigma), 100
mg/ml ascorbic acid (Gibco, Gaithersburg, MD), 100 U/ml each of
penicillin and streptomycin (Irving Scientic, Santa Ana, CA), and
50 mg/ml of L-glutamine (Gibco). AFM experiments were performed
at ambient temperature and atmospheric CO2. To maintain pH, a
commercially available culture medium containing a proprietary
phosphate buffer (Gibco) that can maintain physiologic pH under
atmospheric CO2conditions was used along with the above supple-
ments for the AFM experiments and the corresponding spreading
experiments. No effects on cell spreading from ambient temperature
and atmospheric CO2were observed.
Substrates for cell culture were prepared by incubating 12-mm
glass coverslips overnight with bronectin in a carbonate buffer (15
mM sodium carbonate, 35 mM sodium bicarbonate in water, pH 9.4)
and the nonspecic binding sites were blocked with 1% BSA [21].
Cells were plated at a density of 5000/cm2. For the experiments
studying the effect of actin or myosin, respectively, the drug cytocha-
lasin D (cytoD) at 1
g/ml or the drug ML-9 (1-(5-chloronapthalene-
1-sulfonyl)homopiperazine, HCl), at 7.6
M, was used (both drugs
were from Calbiochem, La Jolla, CA). Stock solutions of the drug
were made in ethanol and contributed a maximum of 0.38% v/v of
ethanol to the nal media volume. At this concentration, ethanol by
itself had no effect on cell shape or stiffness (data not presented). All
AFM measurements were performed at room temperature. Cells
were cultured in ambient air at 37°C until the time of AFM. Cells
exhibited a well-spread morphology by 6 h under these conditions,
similar to those cultured in the presence of 5% CO2.
Atomic force microscopy AFM was performed on a Digital Instru-
ments Multimode Microscope (Digital Instruments, Santa Barbara,
CA) in a uid cell at room temperature by indenting the dorsal
surface of live, adherent cells with a exible cantilever. All measure-
ments were made with standard silicon nitride cantilevers (Digital
Instruments) with a nominal stiffness of 0.06 N/m. Where required,
uid exchange was done by means of a syringe using ports provided
in the uid cell. All force curves were recorded in contact mode. The
AFM was equipped with a Nikon light microscope and video camera,
allowing direct visualization of the cantilever tip on a video monitor.
In this manner the cantilever could be positioned precisely over the
perinuclear region between the spreading edge and the nucleus,
where all measurements were obtained. When performed this way,
all conditions yielded reproducible trends in stiffness.
The recorded force curves were exported to force curve analysis
software (SPMCON) and stiffness was measured as previously de-
scribed [17] using Mathematica software. The method assumes that
the sample deforms upon indentation by a truncated cone, as
F2/
E/1
2兲兴i2tan(
),
where Fis the indentation force, Eis the Young modulus (stiffness)
of the sample,
is its Poissons ratio, iis the sample indentation
(obtained from the force curve data), and
is the half-opening angle
of the indenter, the AFM cantilever tip in this case. Fitting the AFM
force curves to the above equation yields an apparent cell stiffness in
newtons per square meter [17]. Since a cell is expected to be incom-
pressible (i.e., no change in cell volume when indented), the Poisson
ratio was taken to be 0.5. The manufacturers values of 0.06 N/m and
34°were taken for the cantilever stiffness and half-opening angle of
the cone. Due to batch-to-batch variations, the AFM cantilever stiff-
ness can vary from the nominal value. In order to make meaningful
comparisons, all data presented in a gure were acquired with the
same cantilever. It took about 3 min to obtain a force curve on each
cell. Ten force curves, one per cell, on 10 different cells were obtained
representing values at the middle of the time interval for each time
point and the mean stiffness SD was reported.
In the experiments involving the use of cytoskeleton-disrupting
drugs, the drugs at the nal concentration were directly added to
cells in petri dishes 30 min before AFM. After the cells had been
moved to the AFM stage, the drug concentration was maintained
throughout microscopy by perfusing media with the drug through
the AFM uid-cell port using a syringe.
Microscopy and morphometry. Cells were xed at the time points
of interest in 0.5% glutaraldehyde and light microscope images were
obtained on a Zeiss Axiophot25 using Varel optics. The xed cells
were then stained with Coomassie brilliant blue, and spread cell area
3Abbreviations used: ECM, extracellular matrix; AFM, atomic
force microscope, atomic force microscopy; Fn, bronectin; hiFN,
1000 ng/cm2of Fn; loFN, 1 ng/cm2of Fn; cytoD, cytochalasin D;
ML-9, ML-9 hydrochloride, myosin light chain kinase inhibitor;
MLCK, myosin light chain kinase.
93ATOMIC FORCE MICROSCOPY AND CELL STIFFNESS
was obtained by image analysis of digitized light microscope images
[3] using Optimas (Media Cybernetics, Bothell, WA) software. At
least 50 cells were counted per condition and the mean area SD is
reported.
For uorescent actin imaging, cells were cultured on 8-chamber
LabTek slides (Nunc), xed in 1% paraformaldehyde, rinsed in PBS,
and then incubated for 15 min in 0.2% Triton X-100 and 0.1% BSA in
PBS (IF buffer). Rhodamine phalloidin was added at 1
g/ml in IF
buffer for 30 min and then rinsed several times with PBS. Chamber
wells were removed and coverslips were placed onto the glass slides.
Staining was assessed under 100magnication on a Zeiss Axios-
kop uorescence microscope and images were captured using a
Kodak MDS290 digital camera.
DNA synthesis. DNA synthesis was assayed by measuring
[3H]thymidine incorporation. Cells were cultured in Immulon 2B
96-well plates on hiFN as described above. Cytoskeletal drugs were
added at 24 h after plating and [3H]thymidine (ICN, Costa Mesa, CA)
was added at a nal concentration of 10
Ci/ml 48 h after plating.
Parallel plates were used to quantify cell attachment by using the
CyQuant uorimetric assay (Molecular Probes). DNA synthesis was
quantied from incorporated [3H]thymidine as counts/1000 cells per
24 h and values are presented as a percentage of that without any
drugs standard deviation.
RESULTS
Cell Spreading and Stiffness
Stiffness measurements generally involve measur-
ing the deformation of a sample under a dened load.
In such a mode for the AFM, the output is in the form
of a force curve, which plots the sample movement
(moved by a piezoelectric motor) on the x-axis vs the
cantilever deection (or the force exerted by the canti-
lever) on the y-axis. Figure 1 shows two force curves
superimposed, one for a hard surface, glass (curve 1-2-
3), and another for a soft surface, a single hepatocyte
(curve 1-2-4). As the sample is moved toward the can-
tilever (x-axis, moving from right to left with zero de-
ning the point of sample contact with the cantilever),
cantilever deection (y-axis) will be zero as long as the
cantilever does not touch the sample surface. After the
cantilever comes in contact with the sample surface
(point 2 on the gure), it starts to deect upward as the
sample further presses into the cantilever (region 2-3
on glass and region 2-4 on the hepatocyte). The abso-
lute value of the slope for this contact region is unity
for a hard surface, as the cantilever does not indent a
hard surface and is thus deected by as much distance
as the sample moves (region 2-3 in Fig. 1). The absolute
value of the slope is less than unity for a soft surface, as
the cantilever deection is less than the sample move-
ment due to sample indentation by the cantilever (re-
gion 2-4 in Fig. 1). These differences in slope can be
quantied as stiffness in newtons per square meter if a
mechanical model is assumed for the material proper-
ties of the cell, in this case a Hertzian solid [22], as
described under Materials and Methods. It should be
noted that while in certain modes of the AFM it is
possible to make stiffness maps of the cell surface [18,
19], it was not an objective of this study to map the
distribution of cell surface stiffness. It was rather to
study trends in stiffness under conditions that change
cell function. Stiffness measurements in the perinu-
clear region gave reproducible results for the condi-
tions tested.
Asarst step in understanding how hepatocyte stiff-
ness is related to shape, stiffness was measured with
increased spreading. Cells were plated on coverslips
coated with 1000 ng/cm2of bronectin as described
under Materials and Methods. This density of Fn in-
duces signicant cell spreading by 6 h. Stiffness was
measured at time points of interest by placing the
coverslips in the uid cell of the microscope and indent-
ing the dorsal surface of cells with the AFM cantilever
as described above. Cells were imaged in hormonally
dened medium in the absence of any cytoskeleton
disrupting drugs. All measurements were done just
inside the inner boundary of the cell body excluding
both the spreading edge and the nucleus. Area mea-
surements showed that cells increasingly spread for
several hours after plating (Fig. 2A). Stiffness also
increased for several hours after plating (Fig. 2B), mea-
suring 5.34 3.81 kPa, 10.55 7.78 kPa, and 30.3
12.86 kPa at 45 min, 2 h, and 6 h after plating, respec-
tively. The increases were statistically signicant com-
pared to each preceding time point (P0.05, n10
to 14 cells). Previous reports using a different method
have shown a similar relationship between spreading
and indentation stiffness for broblasts [23].
Substrate Density and Stiffness
While Fig. 1 demonstrates a correlation between cell
shape and stiffness, it is not possible to distinguish
between possible effects of increased spreading or sim-
ply increased cell culture and adhesion time. To better
distinguish between the possible effects of cell spread-
ing and culture time, spreading was varied indepen-
FIG. 1. Overview of AFM force curves. Two force curves, one for
a hard surface (curve 1-2-3, on glass) and another for a soft surface
(curve 1-2-4, on a single hepatocyte). The various regions of the force
curve are described in the text. The slope of the force curves (i.e., for
region 2-3 on glass and region 2-4 on the hepatocyte) is different for
the two substrates, reecting differences in their stiffness.
94 BHADRIRAJU AND HANSEN
dent of adhesion time by varying substrate density.
Hepatocytes were plated on a high (1000 ng/cm2)orlow
(1 ng/cm2) density of bronectin and allowed to spread.
After 10 h, the area of hepatocytes on the high density
of bronectin (hiFN) was 1192
m2while the area on
the low density of bronectin (loFN) was 255
m2(Fig.
3A). Stiffness was measured on parallel plates (stiff-
ness of cells on hiFN was measured rst followed by
that of cells on loFN). For corresponding times, the
cells on hiFN, which were more spread, had a stiffness
that was 16.21 5.75 kPa compared to 9.73 6.02 kPa
for cells on loFN (P0.05, n6 to 8 cells; Fig. 3B).
Thus, increased stiffness was independent of culture
time and instead correlated with increased spreading
on a higher substrate density.
Stiffness and the Actomyosin Cytoskeleton
Previous work has shown that changing actin or
myosin activity changes cortical stiffness and that
changes in cell shape are accompanied by changes in
actin arrangement [9] and myosin activity [24] or lo-
calization [25]. Nonmuscle myosin activity, similar to
that of smooth muscle myosin, is regulated by the state
of its phosphorylation. The principal physiological ki-
nase for nonmuscle myosin is myosin light chain ki-
nase (MLCK), which phosphorylates the 20-kDa regu-
latory light chain of myosin [26]. Several previous
studies have shown that perturbing myosin changes
cell stiffness. Myosin-knockout Dictyostelium exhibit
decreased cortical stiffness [27] and 3T3 broblasts
expressing the catalytic domain of myosin light chain
kinase exhibit increased cortical stiffness [28]. Inhibit-
ing myosin light chain kinase activity has also been
shown to change cell shape [29, 30]. Inhibiting myosin
ATPase activity inhibits spreading in broblasts [24].
In addition, myosin cellular localization changes dur-
ing spreading. It is redistributed to the spreading edge
of mouse embryos in vitro [31] and rapidly associates
with polymerized actin in activated platelets as they
change shape [25].
Similar to myosin, disrupting normal actin organi-
zation disrupts cell shape and stiffness in many cell
types. Cytochalasin D, a fungal toxin that inhibits ac-
tin polymerization by binding to the plus end of actin
and preventing it from polymerizing, affects cortical
stiffness and cell shape. Disrupting f-actin integrity
with cytochalasin has previously been shown to reduce
the stiffness of broblasts [19]. In addition to the effect
on stiffness, it has been shown that disrupting the
actin cytoskeleton in chondrocytes disrupts spreading
by both inhibiting the spreading process and causing
spread cells to round up [9, 32, 33]. While the majority
of stiffness studies have been done on either Dictyoste-
lium or stromal cell types such as broblasts and
smooth muscle cells, there is little information avail-
able on the dynamics of cell stiffness in an epithelial
cell type such as hepatocytes. In light of the known
information about the dependence of cortical stiffness
on actomyosin integrity, and the relation between
shape and function, we hypothesized that disrupting
actin and myosin activity would change hepatocyte
shape and stiffness.
Cells were cultured as described under Materials
and Methods. Parallel plates were either xed with
glutaraldehyde for morphometry or assessed by AFM
for stiffness. Cells were photographed using Varel op-
tics to enhance the contrast of the spreading edge, and
arrowheads indicate the location of nuclei. It should be
noted that many hepatocytes are binucleated, as indi-
cated by double arrowheads. CytoD almost completely
inhibited cell spreading when added at the time of
plating (Fig. 4, cytoD-t0). When added to spread cells
5.5 h after plating, there was retraction of the cell
edges and a distinct blebbing of the cell surface within
30 min (Fig. 4, cytoD-t6). To look at the effect of the
drug on spread cells in closer detail, spreading was
quantied upon adding the drug to spread cells. CytoD
FIG. 3. Effect of substrate density on hepatocyte spreading and
stiffness. (A) Hepatocyte spreading on a high (1000 ng/cm2) and low
(1 ng/cm2) density of bronectin. Each value represents the mean cell
area standard deviation of at least 50 cells. (B) Hepatocyte stiff-
ness on a high (1000 ng/cm2) and low (1 ng/cm2) density of bronec-
tin. Each value represents the mean stiffness standard deviation
of 6 to 8 cells.
FIG. 2. Change in hepatocyte spreading and stiffness with time
on a high density (1000 ng/cm2)ofbronectin. (A) Cells were cultured
as described under Materials and Methods and xed with 0.5%
glutaraldehyde. They were then stained with Coomassie blue and
the spread area was measured using digital image analysis. Each
value represents the mean cell area standard deviation of at least
40 cells. (B) Increase in hepatocyte stiffness with increased spread-
ing over time. Each value represents the mean stiffness standard
deviation of 10 to 14 cells.
95ATOMIC FORCE MICROSCOPY AND CELL STIFFNESS
signicantly diminished spread cell area when added
at the time of plating and also when added to cells that
were allowed to spread for 5.5 h and assessed 30 min
later (n61,36 for drug-treated and control cells,
respectively; P0.05, Fig. 5A). To study the corre-
sponding effect on stiffness, AFM was performed on
non-drug-treated cells 6 h after plating and on drug-
treated cells 30 min after drug addition, which was
5.5 h after plating. The addition of cytochalasin D
decreased stiffness by more than half to 38.5% of that
of untreated cells (n9, 7 for drug-treated and control
cells, respectively; P0.05, Fig. 5B).
Since myosin forms an integral part of the actomyo-
sin cytoskeleton, a similar study was done using an
MLCK inhibitor, ML-9, to see the effect of inhibiting
normal myosin activity on hepatocyte shape and stiff-
ness. ML-9 is a specic inhibitor of MLCK with a Ki
(concentration for half-maximal inhibition) of 3.8
M
[34]. While ML-9 can also inhibit protein kinases A and
C, its Kifor MLCK is 8.4 times lower than that for
protein kinase A and 7.1 times lower than that for
protein kinase C [35]. ML-9 (7.6
M) partially inhib-
ited hepatocyte spreading when added at the time of
plating (Fig. 6, ML9-t0). When added to spread cells
5.5 h after plating, there was a distinct cell retraction
within 30 min (Fig. 6, ML9-t6). When spreading was
quantied, ML9 was found to signicantly inhibit
spreading when present throughout the 6-h culture
and to cause signicant cell retraction within 30 min
when added at 5.5 h (n55, 36; P0.05, Fig. 7A).
To look at the corresponding effect on stiffness, AFM
measurements were performed. Unlike in the case of
cytoD, the disruption of myosin light chain phosphor-
ylation using ML-9 increased stiffness more than 3
times to 374% of that of controls (n8 to 10 cells; P
0.05, Fig. 7B).
Actin structure in the presence or absence of cytoD
and ML9 was assessed to ascertain the drugseffects
on the cytoskeleton. Hepatocytes were cultured on
loFN for6horhiFN in the presence or absence of
cytoD and ML9 for either the entire 6-h culture period
or the last 30 min of culture. Cells were xed and
stained with rhodaminephalloidin to visualize the ac-
tin cytoskeleton. Hepatocytes on hiFN for 6 h possess
actin stress bers along the spreading edge which is
notably absent in the rounded cells on loFN (Fig. 8).
ML9 had little effect on actin structure for the 30-min
exposure, but inhibited stress ber formation when
added for the full culture period. CytoD resulted in
disruption of stress bers into a punctate pattern
throughout the cytoplasm when added at either time
(Fig. 8).
Actomyosin Activity and DNA Synthesis
The results presented here demonstrate that dis-
rupting either actin or myosin inhibits cell spreading
in hepatocytes and perturbs cell stiffness. Previous
work had shown a strong correlation between hepato-
cyte spreading and DNA synthesis [3, 8]. Specically, it
has been shown that disrupting f-actin assembly using
cytoD inhibits hepatocyte spreading and DNA synthe-
FIG. 4. Effect of cytochalasin D on hepatocyte morphology. Cells plated on hiFN were treated with 1
g/ml cytochalasin either at the time
of plating (cytoD-t0) or at 5.5 h (cytoD-t6) and xed at 6 h. These are compared to control cells without any drug treatment (no drug).
Arrowheads point to the nucleus. Photos were obtained using Varel optics.
FIG. 5. (A) Effect of cytochalasin D on hepatocyte area. Cells
were plated onto hiFN, treated with 1
M cytochalasin D at the time
of plating (cytoD-t0) or at 5.5 h after plating (cytoD-t6), and xed at
6 h. The spreading is compared to control cells without any drug
treatment (no drug). Cells treated as in Fig. 4 were xed with
glutaraldehyde and stained with Coomassie blue, and cell areas were
quantied by digital image analysis. (B) Effect of cytochalasin D on
hepatocyte stiffness. Cells were treated with 1
g/ml of cytochalasin
D at 5.5 h (cytoD-t6) and stiffness was measured at 6 h. Stiffness is
compared to that of control cells without any drug treatment (no
drug). Each bar is the mean standard deviation of 810 cells for
stiffness measurements.
96 BHADRIRAJU AND HANSEN
sis [3]. Disrupting myosin, which needs f-actin assem-
bly for force generation, has a similar effect on growth
in broblasts [36]. It was hence of interest to study
whether disrupting myosin perturbs hepatocyte
growth. In order to study this, [3H]thymidine incorpo-
ration was compared between 48 and 72 h after plating
in hepatocytes in the presence of 7.6
M ML-9 or 1
g/ml cytochalasin D. Both drugs had the same effect
of inhibiting DNA synthesis. When added at 24 h after
plating, cytoD inhibited hepatocyte DNA synthesis to
12.6% of cells without drug, and ML-9 inhibited it to
22% of cells without drug (Fig. 9). Similar inhibition
was seen when drugs were added at earlier times after
plating (data not shown).
DISCUSSION
This study addresses the relationship between cell
shape changes, stiffness, and the actomyosin cytoskel-
eton. While many previous efforts have focused on in-
vestigating the contribution of actin to cell shape and
stiffness, there have been fewer studies exploring the
same with regard to myosin. The results presented
here show that stiffness increases with cell spreading
in the absence of any drugs. The addition of actin or
myosin disrupting drugs inhibited cell spreading (Figs.
5A and 7A). While both drugs also signicantly
changed stiffness, they had opposite effects. Disrupting
actin assembly decreased cell stiffness to almost a
third of controls (Fig. 5B), whereas inhibiting myosin
light chain kinase activity with the drug ML-9 in-
creased stiffness by more than 3 times (Fig. 7B). Both
drugs also completely inhibited DNA synthesis (Fig. 9).
Hence, these data show for the rst time shape and
stiffness relationships in hepatocytes, a noncontractile,
nonmotile cell type, and present the effect of inhibiting
myosin on hepatocyte growth.
The stiffness of cells in the absence of any drugs was
found to increase with spreading, not only when the
extent of spreading was changed by Fn density (Fig. 3),
but also by allowing cells to spread over time (Fig. 2).
Previous studies with endothelial cells have shown
that integrin bonds formed by RGD-coated magnetic
beads experience a greater resistance to twist in a
magnetic eld at the surface of spread cells on a high
density of bronectin than of round cells on a low
density of bronectin [37]. The experiments presented
here are a different mode of assaying cell stiffness, as
they measure the resistance of cells to indentation
(transverse stiffness). They too show a similar increas-
ing trend in stiffness with spreading. Several lines of
evidence presented below suggest that at least part of
this increase in transverse stiffness with spreading is
due to increased actomyosin contractile forces. In-
creased actomyosin contractile events in muscle bers
can be seen as increased transverse stiffness [38],
showing that changes in cytoskeletal tension can be
detected by indentation measurements. Directly per-
turbing myosin changes cell stiffness, as knocking out
myosin in Dictyostelium results in decreased cortical
stiffness [27]. Further, there is evidence from both the
FIG. 6. Effect of ML-9 on hepatocyte morphology. Cells plated on hiFN were treated with 7.6
M ML-9 either at the time of plating
(ML9-t0) or at 5.5 h (ML9-t6) and xed at 6 h. These are compared to control cells without any drug treatment (no drug). Arrowheads point
to the nucleus. Photos were obtained using Varel optics.
FIG. 7. (A) Effect of ML-9 on hepatocyte area. Cells were treated
with 7.6
M ML-9 at 5.5 h (ML9) and xed at 6 h. They were
subsequently stained with Coomassie blue and morphometry was
done by computerized image analysis. Spreading is compared to
control cells without any drug treatment (no drug). (B) Effect of ML-9
on hepatocyte stiffness. Cells were treated with ML-9 as above and
stiffness was compared to that of control cells without any drug
treatment (no drug). Each bar is the mean standard deviation of
812 cells.
97ATOMIC FORCE MICROSCOPY AND CELL STIFFNESS
literature [24] and our own group (Bhadriraju and
Hansen, manuscript in preparation) to suggest that
myosin ATPase activity increases with cell spreading,
which could be reected in an increased cell stiffness.
The disruption of another major component of the cy-
toskeleton, the microtubule network, has been shown
to not signicantly affect cell stiffness [19]. Put to-
gether, these data suggest that the increase in stiffness
with spreading seen in Figs. 2 and 3 is at least in part
due to actomyosin contractile forces.
To directly test the relative effects of the actin and
myosin cytoskeleton on shape and stiffness, drugs were
used to disrupt their function. Cytochalasin D is an f-
actin disrupter that inhibits spreading in many cell types
including hepatocytes. During this study we found that
ML-9, an inhibitor of myosin light chain kinase, also
inhibits hepatocyte spreading (Fig. 6). Interestingly both
drugs also caused spread cells to round up (Figs. 5A and
7A). Several previous studies have noted rounding of
spread cells upon cytochalasin addition [32, 33, 39]. Since
actin and myosin are together required for myosin gen-
erated force, these results suggest that such forces are
necessary to not only cause cell spreading, as some of the
theories of cell spreading assume, but also to preserve
preformed adhesions. Adhesion bonds that strengthen
upon being stressed have been previously incorporated in
mathematical models [40]. Indeed, there has been direct
experimental evidence to show that integrin bonds be-
come strengthened upon being stressed [41], as might be
expected to happen during spreading, thereby stabilizing
adhesions.
In addition to cell shape, the actomyosin disrupting
drugs also had a signicant effect on stiffness. The
addition of cytoD decreased cell stiffness (Fig. 5B).
CytoD disrupts actin laments by capping the growing
FIG. 8. The effect of actomyosin disrupting drugs on the hepatocyte actin cytoskeleton. Hepatocytes were cultured for 6 h onto 8-chamber
glass slides (LabTek, Nunc) coated with hiFN or loFN. Cytochalasin D (CD) or ML-9 was added to cultures on hiFN for either the last 30 min
of culture (5.56h)orthefull6hofculture (06 h). At 6 h, cells were xed and stained with rhodaminephalloidin. Staining was assessed
under 100magnication.
FIG. 9. The effect of actomyosin disrupting drugs on DNA syn-
thesis in hepatocytes. Cytochalasin (cytoD) or ML-9 was added at
24 h after plating and DNA synthesis was measured by [3H]thymi-
dine incorporation between 48 and 72 h. Values are mean stan-
dard deviation of triplicates from a representative experiment ex-
pressed as the percentage of controls without drug.
98 BHADRIRAJU AND HANSEN
ends. Stiffness studies in other cell types show a de-
crease in stiffness upon cytoD treatment [19, 42, 43]. It
not only disrupts actin lament organization but pre-
sumably also reduces cortical tension as a result of
disrupting myosin association with actin. Unlike the
case of cytoD, ML-9 dramatically increased stiffness
(Fig. 7B) in the absence of any effect on actin morphol-
ogy (Fig. 8), although both drugs inhibited spreading.
These data show that stiffness measurements reveal
changes in the cytoskeleton that are decoupled from
gross cell shape. Further evidence for this is that when
cells were induced to round up by partial trypsiniza-
tion, there was not a statistically signicant change in
stiffness compared to spread cells (data not presented),
unlike in the case of drug-treated cells.
The reason for the increased stiffness upon ML-9 treat-
mentisnotclearalthoughsomepossiblemechanismscan
be speculated upon. Nonmuscle and smooth muscle my-
osin share many common features in their mode of reg-
ulation. Myosin association with actin in nonmuscle and
smooth muscle cells requires light chain phosphorylation
[44]. Actin-associated myosin hydrolyzes ATP to generate
force through cycles of attachment and detachment with
actin laments. At high myosin light chain phosphoryla-
tion levels, cross-bridge detachment is rate-limited by
ADP release and ATP attachment (for the next ATPase
cycle). There are, in general, two conditions under which
a low myosin motor activity is associated with a high
level of stiffness. When ATP is depleted from muscle
bers, myosin cannot detach from actin laments and
goes into a tightly bound state characterized by a large
increase in stiffness. This state, called rigor, is also seen
in nonmuscle cells upon ATP depletion [27]. There is also
considerable evidence to show that the rate of ATPase
cycling is dependent on the extent of myosin light chain
phosphorylation. Myosin association with actin requires
light chain phosphorylation. If actin-associated myosin is
dephosphorylated,this can produce a state of slow cycling
while maintaining force. The stiffness contributed by ac-
tomyosin assemblies is a reection of cross-bridge stiff-
ness. Slow cycling can possibly increase the lifetime of
actomyosin cross-bridges, thereby increasing the mea-
sured stiffness as seen here. It is also possible that ML-9
causes a structural rearrangement of the cytoskeleton
(e.g., bunching up) during cell rounding that shows up as
increased stiffness.
Finally, the fact that inhibiting myosin has as strong
an inhibitory role on DNA synthesis as inhibiting f-actin
does (Fig. 9) points to a key role for normal myosin activ-
ity in hepatocyte growth activation. Previous work had
shown a strong correlation between shape and cell cycle
regulation in several cell types including hepatocytes
[45], capillary endothelial cells [46], and smooth muscle
cells [47]. Previous work also suggests that pathways
involving myosin activation impinge on downstream sig-
nals of the growth pathway including MAP kinase,
p27kip1, and cyclin D1 [36, 46]. In light of the connection
between cell shape and entry into the cell cycle shown
before, and myosin activity and growth presented above,
it is likely that inhibiting myosin might impinge on cell
cycle regulating proteins in hepatocytes. We are cur-
rently investigating the possible role of myosin on cyclin
D1 and MAP kinase regulation.
In conclusion, the results presented here show that
both during spreading and in the presence of actin and
myosin disrupting drugs, hepatocytes undergo changes
in cell stiffness that can be detected by atomic force mi-
croscopy. The data also show that both actin and myosin
are required for hepatocyte spreading and for maintain-
ing the spread shape. Also, the inhibition of hepatocyte
DNA synthesis upon myosin inhibition shows that the
motor protein plays a key role in the growth of a noncon-
tractile, nonmotile cell type. Future studies will focus on
the role of myosin in growth pathways.
The authors thank Kristine Groehler, Lisa Jungers, and Diane
Tobolt for expert technical assistance. This project was supported by
a grant from the University of Minnesota Graduate School and from
the National Science Foundation (MCB9808205).
REFERENCES
1. Stossel, T. P. (1993). On the crawling of animal cells. Science
260, 10861094.
2. McCarthy, J., and Turley, E. A. (1993). Effects of extracellular
matrix components on cell locomotion. Crit. Rev. Oral Biol.
Med. 4, 619637.
3. Mooney, D. J., Hansen, L. K., Vacanti, J. P., Langer, R., Farmer,
S. R., and Ingber, D. E. (1992). Switching from differentiation to
growth in hepatocytes: Control by extracellular matrix. J. Cell.
Phys. 151, 497505.
4. Lee, E. Y.-H., Parry, G., and Bissell, M. J. (1984). Modulation of
secreted proteins of mouse mammary epithelial cells by the
collagenous substrata. J. Cell Biol. 107, 15891598.
5. Chen, C. S., Mrksich, M., Huang, S., Whitesides, G. M., and
Ingber, D. E. (1997). Geometric control of cell life and death.
Science 276, 14251428.
6. Folkman, J., and Moscona, A. (1978). Role of cell shape in
growth control. Nature 346, 760763.
7. Spiegelman, B. M., and Ginty, C. A. (1983). Fibronectin modu-
lation of cell shape and lipogenic gene expression in 3T3adi-
pocytes. Cell 35, 657666.
8. Hansen, L. K., Mooney, D. J., Vacanti, J. P., and Ingber, D. E.
(1994). Integrin binding and cell spreading on extracellular
matrix act at different points in the cell cycle to promote hepa-
tocyte growth. Mol. Biol. Cell. 5, 967975.
9. Mooney, D. J., Langer, R., and Ingber, D. E. (1995). Cytoskel-
etal lament assembly and the control of cell spreading and
function by extracellular matrix. J. Cell Sci. 108, 23112320.
10. Maciver, S. K. (1996). Myosin II function in non-muscle cells.
BioEssays 18, 179182.
11. Binning, G., Quate, C. F., and Gerber, C. (1986). Atomic force
microscope. Phys. Rev. Lett. 56, 930933.
12. Radmacher, M. (1997). Measuring the elastic properties of bio-
logical samples with the AFM. IEEE Eng. Med. Biol. Mag. 16,
4757.
99ATOMIC FORCE MICROSCOPY AND CELL STIFFNESS
13. Domke, J., and Radmacher, M. (1998). Measuring the elastic
properties of thin polymeric lms with the AFM. Langmuir 14,
33203325.
14. Shroff, S. G., Saner, D. R., and Lal, R. (1995). Dynamic micro-
mechanical properties of cultured rat atrial myocytes measured
by atomic force microscopy. Am. J. Physiol. 269, C286292.
15. Laney, D. E., Garcia, R. A., Parsons, S. M., and Hansma, H. G.
(1997). Changes in the elastic properties of cholinergic synaptic
vesicles as measured by atomic force microscopy. Biophys. J.
72, 806812.
16. Radmacher, M., Fritz, M., and Hansma, P. K. (1995). Imaging
soft samples with the atomic force microscope: Gelatin in water
and propanol. Biophys. J. 69, 264270.
17. Rotsch, C., Jacobson, K., and Radmacher, M. (1999). Dimen-
sional and mechanical dynamics of active and stable edges in
motile broblasts investigated by using atomic force micros-
copy. Proc. Natl. Acad. Sci. USA 96, 921926.
18. A-Hassan, E., Heinz, W. F., Antonik, M. D., DCosta, N. P.,
Nageswaran, S., Schoenenberger, C. A., and Hoh, J. H. (1998).
Relative microelastic mapping of living cells by atomic force
microscopy. Biophys. J. 74, 15641578.
19. Rotsch, C., and Radmacher, M. (2000). Drug-induced changes of
cytoskeletal structure and mechanics in broblasts: An atomic
force microscopy study. Biophys. J. 78, 520535.
20. Nyberg, S. L., Shatford, R. A., Payne, W. D., Wu, W. S., and
Cerra, F. B. (1992). Primary culture of rat hepatocytes en-
trapped in cylidrical collagen gels: An in vitro system with
application to the bioarticial liver. Cytotechnology 10, 205.
21. Ingber, D. E. (1990). Fibronectin controls capillary endothelial
cell growth by modulating cell shape. Proc. Natl. Acad. Sci.
USA 87, 35793583.
22. Hertz, H. (1882). U
¨ber die Beru¨hrung fester elastischer Ko˘rper.
Reine Angew. Math. 92, 156171.
23. Elson, E. L. (1988). Cellular mechanics as an indicator of cy-
toskeletal structure and function. [Review] Annu. Rev. Biophys.
Biophys. Chem. 17, 397430.
24. Cramer, L. P., and Mitchison, T. J. (1995). Myosin is involved in
postmitotic cell spreading. J. Cell Biol. 131, 179189.
25. Tanaka, K., Shibata, N., Okamoto, K., Matsusaka, T., Fukuda,
H., Takagi, M., Fujii, N., Toya, N., and Onji, T. (1986). Reorga-
nization of myosin in surface-activated spreading platelets. J.
Ultra. Mol. Struct. R. 97, 165186.
26. Kohama, K., Ye, L. H., Hayakawa, K., and Okagaki, T. (1996).
Myosin light chain kinase: An actin-binding protein that regu-
lates an ATP-dependent interaction with myosin. Trends Phar-
macol. Sci. 17, 284287.
27. Pasternak, C., Spudich, J. A., and Elson, E. L. (1989). Capping
of surface receptors and concomitant cortical tension are gen-
erated by conventional myosin. Nature 341, 549551.
28. Obara, K., Nikcevic, G., Pestic, L., Nowak, G., Lorimer, D. D.,
Guerriero, V. J., Elson, E. L., Paul, R. J., and de Lanerolle, P.
(1995). Fibroblast contractility without an increase in basal
myosin light chain phosphorylation in wild type cells and cells
expressing the catalytic domain of myosin light chain kinase.
J. Biol. Chem. 270, 1873418737.
29. Mobley, P. L., Hedberg, K., Bonin, L., Chen, B., and Grifth, O. H.
(1994). Decreased phosphorylation of four 20-kDa proteins pre-
cedes staurosporine-induced disruption of the actin/myosin cy-
toskeleton in rat astrocytes. Exp. Cell Res. 214, 5566.
30. Miyata, S., Furuya, K., Nakai, S., Bun, H., and Kiyohara, T.
(1999). Morphological plasticity and rearrangement of cytoskel-
etons in pituicytes cultured from adult rat neurohypophysis.
Neurosci. Res. 33, 299306.
31. Sobel, J. S. (1984). Myosin rings and spreading in mouse blas-
tomeres. J. Cell Biol. 99, 11451150.
32. Newman, P., and Watt, F. M. (1988). Inuence of cytochalasin
D-induced changes in cell shape on proteoglycan synthesis by
cultured articular chondrocytes. Exp. Cell Res. 178, 199210.
33. Loty, S., Forest, N., Boulekbache, H., and Sautier, J. M. (1995).
Cytochalasin D induces changes in cell shape and promotes in vitro
chondrogenesis: A morphological study. Biol. Cell 83, 149161.
34. Saitoh, M., Ishikawa, T., Matsushima, S., Naka, M., and
Hidaka, H. (1987). Selective inhibition of catalytic activity of
smooth muscle myosin light chain kinase. J. Biol. Chem. 262,
77967801.
35. Saitoh, M., Naka, M., and Hiroyoshi, H. (1986). The modulatory
role of myosin light chain phosphorylation in human platelet
activation. Biochem. Biophys. Res. Commun. 140, 280287.
36. Cai, S., Pestic-Dragovich, L., ODonnell, M. E., Wang, N., Ing-
ber, D., Elson, E., and De Lanerolle, P. (1998). Regulation of
cytoskeletal mechanics and cell growth by myosin light chain
phosphorylation. Am. J. Physiol. 275, C13491356.
37. Wang, N., and Ingber, D. E. (1994). Control of cytoskeletal
mechanics by extracellular matrix, cell shape, and mechanical
tension. Biophys. J. 66, 21812189.
38. Nyland, L. R., and Maughan, D. W. (2000). Morphology and
transverse stiffness of Drosophila myobrils measured by
atomic force microscopy. Biophys. J. 78, 14901497.
39. Aggeler, J. (1990). Cytoskeletal dynamics in rabbit synovial
broblasts. II. Reformation of stress bers in cells rounded by
treatment with collagenase-inducing agents. Cell Motil. Cy-
toskel. 16, 121132.
40. Dembo, M., Torney, D. C., Saxman, K., and Hammer, D. (1988). The
reaction-limited kinetics of membrane-to-surface adhesion and de-
tachment. Philos. Trans. R Soc. London B Biol. Sci. 234, 5583.
41. Choquet, D., Felsenfeld, D. P., and Sheetz, M. P. (1997). Extra-
cellular matrix rigidity causes strengthening of integrincy-
toskeleton linkages. Cell 88, 3948.
42. Elson, E. L., Pasternak, C., Daily, B., Young, J. I., and McCon-
naughey, W. B. (1984). Cross-linking surface immunoglobulin in-
creases the stiffness of lymphocytes. Mol. Immunol. 21, 12531257.
43. Worthen, G. S., Schwab, B. D., Elson, E. L., and Downey, G. P.
(1989). Mechanics of stimulated neutrophils: Cell stiffening
induces retention in capillaries. Science 245, 183186.
44. Tan, J. L., Ravid, S., and Spudich, J. A. (1992). Control of
nonmuscle myosins by phosphorylation. Annu. Rev. Biochem.
61, 721759.
45. Hansen, L. K., and Albrecht, J. H. (1999). Regulation of the
hepatocyte cell cycle by type I collagen matrix: Role of cyclin D1.
J. Cell Sci. 112, 29712981.
46. Huang, S., Chen, C. S., and Ingber, D. E. (1998). Control of
cyclin D1, p27(Kip1), and cell cycle progression in human cap-
illary endothelial cells by cell shape and cytoskeletal tension.
Mol. Biol. Cell. 9, 31793193.
47. Koyama, H., Raines, E. W., Bornfeldt, K. E., Roberts, J. M., and
Ross, R. (1996). Fibrillar collagen inhibits arterial smooth mus-
cle proliferation through regulation of Cdk2 inhibitors. Cell 87,
10691078.
Received August 20, 2001
Revised version received April 3, 2002
Published online June 20, 2002
100 BHADRIRAJU AND HANSEN
... Filopodia extends the cytoskeleton domain to the surrounding ECM, forcing cells to move forward, and lamellipodia reforms actin expansion towards the cell body itself, which generates a drag flow force that facilitates cell movement [101]. F-actin is known as the hub of mechanotransduction in GBM [50,91,102,103] and is drastically affected by mechanical stress [20]. F-actin can sense the matrix rigidity through integrins connected to focal adhesion sites, where stiffness can intervene to mediate its polymerization ( Figure 5). ...
... Extracellular matrix (ECM) stiffening applies mechanical stress to GBM cells which activates focal adhesion complex formation. Focal adhesion complexes transmit the mechanical stress to cytoskeleton and initiate the CSK remodeling, where F-actin polymerization is highlighted as the hub which orchestrates a metro-system connection between several adaptive cellular signaling pathways [19,20,101,103,[120][121][122]. ...
... Elongated cells have expanded cytoskeletons with stretched morphology, which helps them to move, propagate, and proliferate quickly, interrupting default apoptosis and cell division [124]. [19,20,101,103,[120][121][122]. ...
Article
Full-text available
The biology and physics underlying glioblastoma is not yet completely understood, resulting in the limited efficacy of current clinical therapy. Recent studies have indicated the importance of mechanical stress on the development and malignancy of cancer. Various types of mechanical stress activate adaptive tumor cell responses that include alterations in the extracellular matrix (ECM) which have an impact on tumor malignancy. In this review, we describe and discuss the current knowledge of the effects of ECM alterations and mechanical stress on GBM aggressiveness. Gradual changes in the brain ECM have been connected to the biological and physical alterations of GBM cells. For example, increased expression of several ECM components such as glycosaminoglycans (GAGs), hyaluronic acid (HA), proteoglycans and fibrous proteins result in stiffening of the brain ECM, which alters inter- and intracellular signaling activity. Several mechanosensing signaling pathways have been identified that orchestrate adaptive responses, such as Hippo/YAP, CD44, and actin skeleton signaling, which remodel the cytoskeleton and affect cellular properties such as cell–cell/ECM interactions, growth, and migration/invasion of GBM cells. In vitro, hydrogels are used as a model to mimic the stiffening of the brain ECM and reconstruct its mechanics, which we also discuss. Overall, we provide an overview of the tumor microenvironmental landscape of GBM with a focus on ECM stiffening and its associated adaptive cellular signaling pathways and their possible therapeutic exploitation.
... Cytochalasin inhibits actin polymerization, presumably by binding with high affinity to the barbed ends of actin filaments (F-actin) and preventing the addition of monomers (G-actin) to these sites [17]. Thus cytochalasin reduces cell stiffness by disrupting the actin cytoskeleton [18,19]. In each experiment, cytochalasin was administered at a concentration of 2 µM. ...
... All images were captured using a microscope (Olympus, Tokyo, Japan) at an excitation wavelength of 488-nm with a 100-ms exposure time. Schaub et al. reported that the fluorescence intensity of phalloidin-stained images is proportional to the density of actin filaments and that variations in staining intensity reflect variations in actin density [18]. We used Image J software (NIH Image, Bethesda, MD, USA) to measure the mean intensity of F-actin staining [18]. ...
... Schaub et al. reported that the fluorescence intensity of phalloidin-stained images is proportional to the density of actin filaments and that variations in staining intensity reflect variations in actin density [18]. We used Image J software (NIH Image, Bethesda, MD, USA) to measure the mean intensity of F-actin staining [18]. To measure intensity variations, we used the ratio of standard deviation (SD) of intensity to the mean intensity of the image. ...
Article
Full-text available
Osteosarcoma is the most common primary malignant bone tumor. The cause of death due to osteosarcoma is typically a consequence of metastasis to the lung. Controlling metastasis leads to improved prognosis for osteosarcoma patients. The cell stiffness of several tumor types is involved in metastatic potential; however, it is unclear whether the metastatic potential of osteosarcoma depends on cell stiffness. In this study, we analyzed the cell stiffness of the low metastatic Dunn cell line and its highly metastatic LM8 subline, and compared actin organization, cell proliferation, and metastasis. Actin cytoskeleton, polymerization, stiffness, and other cellular properties were analyzed. The organization of the actin cytoskeleton was evaluated by staining F-actin with Alexa Fluor 488 phalloidin. Cell stiffness was measured using Atomic Force Microscopy (AFM). Cell proliferation, migration, invasion, and adhesion were also evaluated. All experiments were performed using mouse osteosarcoma cell lines cultured in the absence and presence of cytochalasin. In LM8 cells, actin polymerization was strongly suppressed and actin levels were significantly lower than in Dunn cells. Stiffness evaluation revealed that LM8 cells were significantly softer than Dunn. Young’s modulus images showed more rigid fibrillar structures were present in Dunn cells than in LM8 cells. LM8 cells also exhibited a significantly higher proliferation. The migration and invasion potential were also higher in LM8 cells, whereas the adhesion potential was higher in Dunn cells. The administration of cytochalasin resulted in actin filament fragmentation and decreased actin staining intensity and cell stiffness in both LM8 and Dunn cells. Cells with high metastatic potential exhibited lower actin levels and cell stiffness than cells with low metastatic potential. The metastatic phenotype is highly correlated to actin status and cell stiffness in osteosarcoma cells. These results suggest that evaluation of actin dynamics and cell stiffness is an important quantitative diagnostic parameter for predicting metastatic potential. We believe that these parameters represent new reliable quantitative indicators that can facilitate the development of new drugs against metastasis.
... F-actin in ES-2 cells maintains a simple ring-like structure with a lower density. Previously, studies showed that higher absolute levels of F-actin are associated with increased cell stiffness, whereas reductive or low-density F-actin correlates with lower cell stiffness [26][27][28]. Meanwhile, the transcriptional levels of actinin alpha 3 (ACTN3) and Myosin 1A (MYO1A) genes, which increase the cell stiffness, were also significantly lower in naïve ES-2 cells than that in naïve SKOV3 cells [29,30] (Figure 3A). ...
... Myosin II hydrolyzes ATP to generate force through attachment and detachment with F-actin filaments. A high myosin II activity and low density of the cross-linked F-actin filament are associated with a low level of cell stiffness [26,39]. In this study, blocking the myosin II by blebbistatin significantly increased the cell stiffness of ovarian cancer cells, as well as enhanced sensitivity of ES-2 cells to SEC2/ST-4-induced immune cytotoxicity. ...
Article
Full-text available
As a biological macromolecule, the superantigen staphylococcal enterotoxin C2 (SEC2) is one of the most potent known T-cell activators, and it induces massive cytotoxic granule production. With this property, SEC2 and its mutants are widely regarded as immunomodulating agents for cancer therapy. In a previous study, we constructed an MHC-II-independent mutant of SEC2, named ST-4, which exhibits enhanced immunocyte stimulation and antitumor activity. However, tumor cells have different degrees of sensitivity to SEC2/ST-4. The mechanisms of immune resistance to SEs in cancer cells have not been investigated. Herein, we show that ST-4 could activate more powerful human lymphocyte granule-based cytotoxicity than SEC2. The results of RNA-seq and atomic force microscopy (AFM) analysis showed that, compared with SKOV3 cells, the softer ES-2 cells could escape from SEC2/ST-4-induced cytotoxic T-cell-mediated apoptosis by regulating cell softness through the CDC42/MLC2 pathway. Conversely, after enhancing the stiffness of cancer cells by a nonmuscle myosin-II-specific inhibitor, SEC2/ST-4 exhibited a significant antitumor effect against ES-2 cells by promoting perforin-dependent apoptosis and the S-phase arrest. Taken together, these data suggest that cell stiffness could be a key factor of resistance to SEs in ovarian cancer, and our findings may provide new insight for SE-based tumor immunotherapy.
... Complex biological processes of cancer cells are associated with well-described pathways (e.g., cell adhesion, cytoskeletal remodeling, EMT, and the nucleoskeleton) (Bhadriraju and Hansen, 2002;de las Heras et al., 2013;Ribatti et al., 2020). Conversely, the molecular mechanisms that regulate CTC mechanics in the microcirculation are rather elusive. ...
... Intermediate filaments are important for resistance to cell deformation in response to large strains and operate as a safety net against severe distortion that would damage the cell (Charrier and Janmey, 2016). Therefore, the cytoskeleton is crucial for the cell stiffness and capacity to passively deform, and also for the general cell shape actively adopted during various activities (Bhadriraju and Hansen, 2002). The cytoskeleton also places the cell organelles inside its network of filaments, providing a direct connection between cell shape/ deformation and important organelle repositioning during the metastatic cascade. ...
Article
Full-text available
Metastatic progression is the deadliest feature of cancer. Cancer cell growth, invasion, intravasation, circulation, arrest/adhesion and extravasation require specific mechanical properties to allow cell survival and the completion of the metastatic cascade. Circulating tumor cells (CTCs) come into contact with the capillary bed during extravasation/intravasation at the beginning of the metastatic cascade. However, CTC mechanobiology and survival strategies in the bloodstream, and specifically in the microcirculation, are not well known. A fraction of CTCs can extravasate and colonize distant areas despite the biomechanical constriction forces that are exerted by the microcirculation and that strongly decrease tumor cell survival. Furthermore, accumulating evidence shows that several CTC adaptations, via molecular factors and interactions with blood components (e.g., immune cells and platelets inside capillaries), may promote metastasis formation. To better understand CTC journey in the microcirculation as part of the metastatic cascade, we reviewed how CTC mechanobiology and interaction with other cell types in the bloodstream help them to survive the harsh conditions in the circulatory system and to metastasize in distant organs.
... One of the most likely explanations for this effect is the dependence of sonoporation efficacy on the biophysical characteristics of the cell. Some articles suggested that cell cycle progression is accompanied by cytoskeleton reorganization that significantly changes cell mechanical properties [89,90]. To confirm this, Fan et al. evaluated single-cell responses to sonoporation of HeLa cells synchronized in a particular cycle phase. ...
Article
Full-text available
Sonoporation is the process of transient pore formation in the cell membrane triggered by ultrasound (US). Numerous studies have provided us with firm evidence that sonoporation may assist cancer treatment through effective drug and gene delivery. However, there is a massive gap in the body of literature on the issue of understanding the complexity of biophysical and biochemical sonoporation-induced cellular effects. This study provides a detailed explanation of the US-triggered bioeffects, in particular, cell compartments and the internal environment of the cell, as well as the further consequences on cell reproduction and growth. Moreover, a detailed biophysical insight into US-provoked pore formation is presented. This study is expected to review the knowledge of cellular effects initiated by US-induced sonoporation and summarize the attempts at clinical implementation.
... Cancer cell stiffness has been reported to correlate well with the disease invasiveness, due to cellular stiffness variations in tumors following changes in cell cytoskeleton, and membrane microviscosity, [6,8,[11][12][13]. Metastatic cells have elastic features that allow them to detach from the primary tumor, penetrate the walls of lymphatic or blood vessels, and create secondary or metastatic tumors [14][15][16][17]. Thus, cancer cell stiffness and its associated properties could form a diagnosis tool, by classifying cancer cell types for early detection, monitoring, and development of specific cancer treatment [18]. ...
Article
Full-text available
We present an analysis method that can automatically classify live cancer cells from cell lines based on a small data set of quantitative phase imaging data without cell staining. The method includes spatial image analysis to extract the cell phase spatial fluctuation map, derived from the quantitative phase map of the cell measured without cell labeling, thus without prior knowledge on the biomarker. The spatial fluctuations are indicative of the cell stiffness, where cancer cells change their stiffness as cancer progresses. In this paper, the quantitative phase spatial fluctuations are used as the basis for a deep-learning classifier for evaluating the cell metastatic potential. The spatial fluctuation analysis performed on the quantitative phase profiles before inputting them to the neural network was proven to increase the classification results in comparison to inputting the quantitative phase profiles directly, as done so far. We classified between primary and metastatic cancer cells and obtained 92.5% accuracy, in spite of using a small training set, demonstrating the method potential for objective automatic clinical diagnosis of cancer cells in vitro .
Preprint
Full-text available
Protein aggregation is a common underlying feature of neurodegenerative disorders. Cells expressing neurodegeneration associated mutant proteins show altered uptake of ligands, suggestive of impaired endocytosis, in a manner as yet unknown. Using live cell imaging, we show that clathrin mediated endocytosis (CME) is affected due to altered actin cytoskeletal organization in the presence of Huntingtin aggregates. Additionally, we find that cells containing Huntingtin aggregates are stiffer and less viscous than their wild type counterparts due to altered actin conformation, and not merely due to the physical presence of aggregate(s). We further demonstrate that CME and cellular viscosity can be rescued by overexpressing Hip1, Arp2/3 or transient LatrunculinA treatment. Examination of other pathogenic aggregates revealed that only a subset of these display defective CME, along with altered actin organization and increased stiffness. Together, our results point to an intimate connection between functional CME, actin organization and cellular stiffness in the context of neurodegeneration.
Article
During tumorigenesis, the mechanical properties of cancer cells change markedly, with decreased stiffness often accompanying a more invasive phenotype. Less is known about the changes in mechanical parameters at intermediate stages in the process of malignant transformation. We have recently developed a pre-tumoral cell model by stably transducing the immortalized but non-tumorigenic human keratinocyte cell line HaCaT with the E5, E6 and E7 oncogenes from HPV-18, one of the leading causes of cervical cancer and other types of cancer worldwide. We have used atomic force microscopy (AFM) to measure cell stiffness and to obtain mechanical maps of parental HaCaT and HaCaT E5/E6/E7-18 cell lines. We observed a significant decrease in Young's modulus in HaCaT E5/E6/E7-18 cells measured by nanoindentation in the central region, as well as decreased cell rigidity in regions of cell-cell contact measured by Peakforce Quantitative Nanomechanical Mapping (PF-QNM). As a morphological correlate, HaCaT E5/E6/E7-18 cells displayed a significantly rounder cell shape than parental HaCaT cells. Our results therefore show that decreased stiffness with concomitant perturbations in cell shape are early mechanical and morphological changes during the process of malignant transformation.
Article
Alternatively activated or M2 macrophages, as opposed to the well characterized pro-inflammatory or M1 macrophages, vitally regulate anti-inflammation, wound healing, and tissue repair to maintain tissue homeostasis. Although ubiquitous presence of macrophages in diverse tissues, exposed to different physical environments, infers distinct immune responses of M2 macrophages with high phenotypic heterogeneity, the underlying mechanism of how the varying extracellular mechanical conditions alter their immunological activation remains unclear. Here, we demonstrate that M2 activation requires a threshold mechanical cue from the extracellular microenvironment, and matrix rigidity-dependent macrophage spreading is mediated by the F-actin formation that is essential to regulate mechanosensitive M2 activation of macrophages. We identified a new mechanosensing function of STAT6 (signal transducer and activator of transcription 6), a key transcription factor for M2 activation, whose intranuclear transportation is promoted by the rigid matrix that facilitates the F-actin formation. Our findings further highlight the critical role of mechanosensitive M2 activation of macrophages in long-term adaptation to the extracellular microenvironment by bridging nuclear mechanosensation and immune responses.
Article
The study of physical and mechanical features of cancer cells, or cancer cell mechanobiology, is a new frontier in cancer research. Such studies may enhance our understanding of the disease process, especially mechanisms associated with cancer cell invasion and metastasis, and may help the effort of developing diagnostic biomarkers and therapeutic drug targets. Cancer cell mechanobiological changes are associated with the complex interplay of activation/inactivation of multiple signaling pathways, which can occur at both the genetic or epigenetic level, and the interactions with the cancer microenvironment. It has been shown that metastatic tumor cells are more compliant than morphologically similar benign cells in actual human samples. Subsequent studies from us and others further demonstrated that cell mechanical properties are strongly associated with cancer cell invasive and metastatic potential, and thus may serve as a diagnostic marker of detecting cancer cells in human body fluid samples. In this review, we will provide a brief narrative of the molecular mechanisms underlying cancer cell mechanobiology, the technological platforms utilized to study cancer cell mechanobiology, the status of cancer cell mechanobiological studies in various cancer types, and the potential clinical applications of cancer cell mechanobiological study in cancer early detection, diagnosis, and treatment.
Article
Full-text available
The scanning tunneling microscope is proposed as a method to measure forces as small as 10-18 N. As one application for this concept, we introduce a new type of microscope capable of investigating surfaces of insulators on an atomic scale. The atomic force microscope is a combination of the principles of the scanning tunneling microscope and the stylus profilometer. It incorporates a probe that does not damage the surface. Our preliminary results in air demonstrate a lateral resolution of 30 ÅA and a vertical resolution less than 1 Å.
Article
Full-text available
The spatial and temporal changes of the mechanical properties of living cells reflect complex underlying physiological processes. Following these changes should provide valuable insight into the biological importance of cellular mechanics and their regulation. The tip of an atomic force microscope (AFM) can be used to indent soft samples, and the force versus indentation measurement provides information about the local viscoelasticity. By collecting force-distance curves on a time scale where viscous contributions are small, the forces measured are dominated by the elastic properties of the sample. We have developed an experimental approach, using atomic force microscopy, called force integration to equal limits (FIEL) mapping, to produce robust, internally quantitative maps of relative elasticity. FIEL mapping has the advantage of essentially being independent of the tip-sample contact point and the cantilever spring constant. FIEL maps of living Madine-Darby canine kidney (MDCK) cells show that elasticity is uncoupled from topography and reveal a number of unexpected features. These results present a mode of high-resolution visualization in which the contrast is based on the mechanical properties of the sample.
Article
It has been shown previously that cultures of mouse mammary epithelial cells retain their characteristic morphology and their ability to produce gamma-casein, a member of the casein gene family, only if they are maintained on floating collagen gels (Emerman, J.T., and D.R. Pitelka, 1977, In Vitro, 13:316-328). In this paper we show: (a) Cells on floating collagen gels secrete not only gamma-casein but also alpha 1-, alpha 2-, and beta-caseins. These are not secreted by cells on plastic and are secreted to only a very limited extent by cells on attached collagen gels. (b) The floating collagen gel regulates at the level of synthesis and/or stabilization of the caseins rather than at the level of secretion alone. Contraction of the floating gel is important in that cells cultured on floating glutaraldehyde cross-linked gels do not secrete any of the caseins. (c) The secretion of an 80,000-mol-wt protein, most probably transferrin, and a 67,000-mol-wt protein, probably butyrophilin, a major protein of the milk fat globule membrane are partially modulated by substrata. However, in contrast to the caseins, these are always detectable in media from cells cultured on plastic and attached gels. (d) Whey acidic protein, a major whey protein, is actively secreted by freshly isolated cells but is secreted in extremely limited quantities in cultured cells regardless of the nature of the substratum used. alpha-Lactalbumin secretion is also decreased significantly in cultured cells. (e) A previously unreported set of proteins, which may be minor milk proteins, are prominently secreted by the mammary cells on all substrata tested. We conclude that while the substratum profoundly influences the secretion of the caseins, it does not regulate the expression of every milk-specific protein in the same way. The mechanistic implications of these findings are discussed.
Article
The atomic force microscope (AFM) is a promising new method for studying the surface structure of both conductors and insulators. In mapping a graphite surface with an insulating stylus, we have achieved a resolution better than 2.5 Å.
Article
The elastic properties of thin gelatin films were investigated with the atomic force microscope (AFM). The degree of swelling and thus the softness of the gelatin can be tuned by immersing it in mixtures of propanol and water. Therefore, we have chosen gelatin films as a model system to characterize the measurement of elasticity of thin and soft samples. The major aim of this study was to investigate the influence of the film thickness on the apparent elastic (Young's) modulus. Thus, we prepared wedge-shaped samples with a well-defined thickness of up to 1 mu m. The Young's modulus of our samples was between 1 MPa and 20 kPa depending on the degree of swelling. The elasticity was calculated by analyzing the recorded force curves with the help of the Hertz model. We show that the calculated Young's modulus is dependent on the local film thickness and the applied loading force of the AFM tip. Thus, the influence of the hard substrate on the calculated softness of the film can be characterized as a function of indentation. It was possible to determine the elastic properties of gelatin films with a thickness down to 50 nm and a Young's modulus of similar to 20 kPa.
Article
Cholinergic synaptic vesicles from Torpedo californica have been probed with the atomic force microscope in aqueous buffers to map and measure their elastic properties. Elastic properties were mapped with a new atomic force microscope technique known as force mapping. Force mapping of vesicles showed that the centers of the vesicles are harder or stiffer than the peripheral areas in the three buffers that were investigated. These were an isoosmotic buffer, a hypoosmotic buffer, and an isoosmotic buffer with 5 mM CaCl2 added. The hardness of the vesicular centers was quantified by calculation of the elastic modulus. Elastic moduli were in the range of 2-13 × 105 Pa. Vesicular centers were hardest in calcium-containing buffer and softest in isoosmotic buffer. Hypotheses are presented for the composition and function of the hard centers.