+ All Categories
Home > Documents > Biophysical Regulation of Histone Acetylation in Mesenchymal Stem Cells

Biophysical Regulation of Histone Acetylation in Mesenchymal Stem Cells

Date post: 25-Nov-2016
Category:
Upload: song
View: 215 times
Download: 2 times
Share this document with a friend
8
Biophysical Regulation of Histone Acetylation in Mesenchymal Stem Cells Yuan Li, †‡ Julia S. Chu, Kyle Kurpinski, Xian Li, †‡ Diana M. Bautista, § Li Yang, * K.-L. Paul Sung, { and Song Li * Bioengineering College, Chongqing University, Chongqing, China; Department of Bioengineering and § Department of Molecular and Cell Biology, University of California, Berkeley, California; and { Departments of Bioengineering and Orthopaedics, University of California, San Diego, La Jolla, California ABSTRACT Histone deacetylation and acetylation are catalyzed by histone deacetylase (HDAC) and histone acetyltransfer- ase, respectively, which play important roles in the regulation of chromatin remodeling, gene expression, and cell functions. However, whether and how biophysical cues modulate HDAC activity and histone acetylation is not well understood. Here, we tested the hypothesis that microtopographic patterning and mechanical strain on the substrate regulate nuclear shape, HDAC activity, and histone acetylation. Bone marrow mesenchymal stem cells (MSCs) were cultured on elastic membranes patterned with parallel microgrooves 10 mm wide that kept MSCs aligned along the axis of the grooves. Compared with MSCs on an unpatterned substrate, MSCs on microgrooves had elongated nuclear shape, a decrease in HDAC activity, and an increase of histone acetylation. To investigate anisotropic mechanical sensing by MSCs, cells on the elastic micropatterned membranes were subjected to static uniaxial mechanical compression or stretch in the direction parallel or perpendicular to the microgrooves. Among the four types of loads, compression or stretch perpendicular to the microgrooves caused a decrease in HDAC activity, accompanied by the increase in histone acetylation and slight changes of nuclear shape. Knocking down nuclear matrix protein lamin A/C abolished mechanical strain-induced changes in HDAC activity. These results demonstrate that micro- pattern and mechanical strain on the substrate can modulate nuclear shape, HDAC activity, and histone acetylation in an aniso- tropic manner and that nuclear matrix mediates mechanotransduction. These findings reveal a new mechanism, to our knowledge, by which extracellular biophysical signals are translated into biochemical signaling events in the nucleus, and they will have significant impact in the area of mechanobiology and mechanotransduction. INTRODUCTION Bone marrow mesenchymal stem cells (MSCs) can be expanded and stimulated to differentiate into a variety of cell types (1–4), demonstrating the potential of MSCs as a possible cell source for stem cell therapy. However, the effects of microenvironmental cues on MSCs are not well understood. MSCs have been used for the regeneration of tissues in blood vessels, heart, cartilage, and bone (2,3,5). The cells in all of these tissues are subjected to mechanical strain in the extracellular matrix (ECM) due to external mechanical loads. We have shown that MSCs can sense the direction of uniaxial mechanical stretch and respond differently in gene expression (6), but the underlying mech- anisms remain to be determined. Besides the transcriptional regulation by transcriptional factors, gene expression can be turned on or off by changes in chromatin structure, which are modulated by a number of posttranslational modifications at the amino-terminal tails of nucleosomal histones, including acetylation/deacetyla- tion, phosphorylation, methylation, and adenosine diphos- phate (ADP) ribosylation (7). The acetylation status of chromatin is dynamic, balanced by the activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs) (8), which can activate or suppress the expression of specific genes (9). In addition, nuclear matrix, locating at the interface between cytoplasm and nucleus, not only functions as a structural framework but is also involved in the regulation of chromosome and gene expression (10,11). For example, lamin A/C deficiency causes conden- sation and reorganization of chromatin (12). There is evidence that biophysical factors may regulate cell shape, nuclear shape, and histone acetylation. It has been reported that smooth muscle cells (SMCs) on micro- patterned substrates have elongated cell shape and nuclear shape, correlated with a lower proliferation rate of SMCs compared with SMCs in a spread-out morphology (13). In 3D cultures as well as on nonadhesive and micropatterned substrata, mammary epithelial cell rounding induced deace- tylation of histones H3 and H4 in the absence of biochem- ical cues (14). Regarding the potential role of nuclear matrix in mechanotransduction, a previous study shows that lamin A/C deficiency inhibits the gene expression of egr-1 and iex-1 induced by cyclic biaxial strain (15). However, it is not clear whether nuclear shape and mechan- ical factors modulate histone acetylation, and the role of nuclear matrix in mechanotransduction is not well understood. We postulated that both substrate and mechanical loading could exert direct effects on cell nuclei, chromatin remodel- ing and gene expression. To test this possibility, we used micropatterning techniques to fabricate microgrooves on poly(dimethyl siloxane) (PDMS) membranes to control cell morphology and alignment. We demonstrated that HDAC activity was decreased and the level of acetylated- histone H3 (AcH3) was increased in nucleus when cells Submitted October 5, 2010, and accepted for publication March 9, 2011. *Correspondence: [email protected] or [email protected] Editor: Denis Wirtz. Ó 2011 by the Biophysical Society 0006-3495/11/04/1902/8 $2.00 doi: 10.1016/j.bpj.2011.03.008 1902 Biophysical Journal Volume 100 April 2011 1902–1909
Transcript

1902 Biophysical Journal Volume 100 April 2011 1902–1909

Biophysical Regulation of Histone Acetylation in Mesenchymal Stem Cells

Yuan Li,†‡ Julia S. Chu,‡Kyle Kurpinski,‡Xian Li,†‡Diana M. Bautista,§Li Yang,†*K.-L. Paul Sung,†{and Song Li‡*†Bioengineering College, Chongqing University, Chongqing, China; ‡Department of Bioengineering and §Department of Molecular and CellBiology, University of California, Berkeley, California; and {Departments of Bioengineering and Orthopaedics, University of California,San Diego, La Jolla, California

ABSTRACT Histone deacetylation and acetylation are catalyzed by histone deacetylase (HDAC) and histone acetyltransfer-ase, respectively, which play important roles in the regulation of chromatin remodeling, gene expression, and cell functions.However, whether and how biophysical cues modulate HDAC activity and histone acetylation is not well understood. Here,we tested the hypothesis that microtopographic patterning and mechanical strain on the substrate regulate nuclear shape,HDAC activity, and histone acetylation. Bone marrow mesenchymal stem cells (MSCs) were cultured on elastic membranespatterned with parallel microgrooves 10 mm wide that kept MSCs aligned along the axis of the grooves. Compared withMSCs on an unpatterned substrate, MSCs on microgrooves had elongated nuclear shape, a decrease in HDAC activity, andan increase of histone acetylation. To investigate anisotropic mechanical sensing by MSCs, cells on the elastic micropatternedmembranes were subjected to static uniaxial mechanical compression or stretch in the direction parallel or perpendicular to themicrogrooves. Among the four types of loads, compression or stretch perpendicular to the microgrooves caused a decrease inHDAC activity, accompanied by the increase in histone acetylation and slight changes of nuclear shape. Knocking down nuclearmatrix protein lamin A/C abolished mechanical strain-induced changes in HDAC activity. These results demonstrate that micro-pattern and mechanical strain on the substrate can modulate nuclear shape, HDAC activity, and histone acetylation in an aniso-tropic manner and that nuclear matrix mediates mechanotransduction. These findings reveal a new mechanism, to ourknowledge, by which extracellular biophysical signals are translated into biochemical signaling events in the nucleus, andthey will have significant impact in the area of mechanobiology and mechanotransduction.

INTRODUCTION

Bone marrow mesenchymal stem cells (MSCs) can beexpanded and stimulated to differentiate into a variety ofcell types (1–4), demonstrating the potential of MSCs asa possible cell source for stem cell therapy. However, theeffects of microenvironmental cues on MSCs are not wellunderstood. MSCs have been used for the regeneration oftissues in blood vessels, heart, cartilage, and bone (2,3,5).The cells in all of these tissues are subjected to mechanicalstrain in the extracellular matrix (ECM) due to externalmechanical loads. We have shown that MSCs can sensethe direction of uniaxial mechanical stretch and responddifferently in gene expression (6), but the underlying mech-anisms remain to be determined.

Besides the transcriptional regulation by transcriptionalfactors, gene expression can be turned on or off by changesin chromatin structure, which are modulated by a number ofposttranslational modifications at the amino-terminal tailsof nucleosomal histones, including acetylation/deacetyla-tion, phosphorylation, methylation, and adenosine diphos-phate (ADP) ribosylation (7). The acetylation status ofchromatin is dynamic, balanced by the activities of histoneacetyltransferases (HATs) and histone deacetylases(HDACs) (8), which can activate or suppress the expressionof specific genes (9). In addition, nuclear matrix, locating atthe interface between cytoplasm and nucleus, not only

Submitted October 5, 2010, and accepted for publication March 9, 2011.

*Correspondence: [email protected] or [email protected]

Editor: Denis Wirtz.

� 2011 by the Biophysical Society

0006-3495/11/04/1902/8 $2.00

functions as a structural framework but is also involved inthe regulation of chromosome and gene expression(10,11). For example, lamin A/C deficiency causes conden-sation and reorganization of chromatin (12).

There is evidence that biophysical factors may regulatecell shape, nuclear shape, and histone acetylation. It hasbeen reported that smooth muscle cells (SMCs) on micro-patterned substrates have elongated cell shape and nuclearshape, correlated with a lower proliferation rate of SMCscompared with SMCs in a spread-out morphology (13). In3D cultures as well as on nonadhesive and micropatternedsubstrata, mammary epithelial cell rounding induced deace-tylation of histones H3 and H4 in the absence of biochem-ical cues (14). Regarding the potential role of nuclearmatrix in mechanotransduction, a previous study showsthat lamin A/C deficiency inhibits the gene expression ofegr-1 and iex-1 induced by cyclic biaxial strain (15).However, it is not clear whether nuclear shape and mechan-ical factors modulate histone acetylation, and the role ofnuclear matrix in mechanotransduction is not wellunderstood.

We postulated that both substrate and mechanical loadingcould exert direct effects on cell nuclei, chromatin remodel-ing and gene expression. To test this possibility, we usedmicropatterning techniques to fabricate microgrooves onpoly(dimethyl siloxane) (PDMS) membranes to controlcell morphology and alignment. We demonstrated thatHDAC activity was decreased and the level of acetylated-histone H3 (AcH3) was increased in nucleus when cells

doi: 10.1016/j.bpj.2011.03.008

Biophysical Regulation of HDAC 1903

and nuclei were elongated on micropatterned surfaces. Witha precise control of cell alignment and the direction ofuniaxial mechanical strain, we showed that compressionand stretch in the direction perpendicular to cell alignmentinduced nuclear shape change, a decrease of HDAC activity,and an increase in histone acetylation. Furthermore,nuclear-matrix lamin mediated mechanotransductionleading to HDAC activity change in response to mechanicalstrain. These results illustrate that micropatterned substrateand mechanical loading can modulate nuclear shape, HDACactivity, and histone acetylation in an anisotropic andnuclear-matrix-dependent manner, which offers a newmechanism, to our knowledge, by which biophysical factorsregulate cell functions.

MATERIALS AND METHODS

Microfabrication and soft lithography

To create patterned membranes with parallel microgrooves (10 mm in width

and 3 mm in height, with 10 mm distance between each groove), we used

microfabrication techniques described previously (6). Briefly, photoresist

(OIR 897-10I, Arch Chemicals, Norwalk, CT) was spin-coated onto

a silicon wafer, and a patterned photomask was used to expose the photo-

resist to ultraviolet light. After washing away the unpolymerized photore-

sist, PDMS was prepared according to the manufacturer protocol

(Sylgard 184, Dow Corning, Midland, MI), spin-coated onto the patterned

silicon wafers to desired thickness (~250 mm), degassed under vacuum, and

cured at 70�C for 2 h. The resulting micropatterned membranes were

removed from the template, cut to appropriate dimensions (for assembly

into custom-built stretch chambers), and thoroughly washed and sonicated

before use. The surface topography of micropatterned PDMS membranes

was examined by scanning electron microscopy (SEM) (13). In this study,

we used 10-mm wide microgrooves to control cell and nuclear shape, as our

preliminary studies showed that 30–50 mm microgrooves did not consis-

tently induce changes in nuclear shape.

Cell culture

Human MSCs were acquired from Cambrex (East Rutherford, NJ) and

cultured in MSC growth medium (MSCGM) (Cambrex) for expansion

without differentiation. After cell expansion, MSCs were subjected to

flow cytometry analysis of surface marker expression (positive for

CD105, CD166, CD29, and CD44, and negative for CD34, CD14, and

CD45) and differentiation assays (smooth muscle, osteogenic, and chondro-

genic lineages) to confirm the undifferentiated state of MSCs. Cells were

kept in a humidified incubator at 37�C and supplemented with 5% CO2.

Applying uniaxial static strain

For mechanical stretch or compression experiments, micropatterned

membranes were O2 plasma-treated with a Plasma-PrepTM II plasma

etcher (Structure Probe, West Chester, PA) for 1 min and coated with 2%

gelatin to support cell adhesion on the membranes. Under the experimental

conditions, serum proteins (e.g., fibronectin) and matrix proteins synthe-

sized by cells could also adsorb on the surfaces. Membranes were assem-

bled into custom-built uniaxial stretch chambers with microgrooves

oriented either parallel or perpendicular to the axis of strain, as stated for

each experiment. MSCs were seeded at 70% confluency on the patterned

surfaces in a medium containing Dulbecco’s modified Eagle’s medium,

10% fetal bovine serum, 1% penicillin/streptomycin antibiotic mix, and

1% fungizone. Cell culture was maintained in a humidified incubator at

37�C and supplemented with 5% CO2.

For samples subjected to uniaxial stretch, the PDMS membranes were

assembled taut. Cells were then seeded on the membranes and allowed to

fully attach. After 1 day in culture, the membranes were subjected to

20% uniaxial stretch. For samples subjected to uniaxial compression, the

PDMS membranes were first assembled in a 20% prestretched configura-

tion. Cells were then seeded onto these prestretched membranes and

allowed to fully attach. After 1 day in culture, the membranes were released

to 0% strain, which was equivalent to 20% compression for the attached

cells. For controls, cells were seeded on taut but unstrained membranes,

and no loads were applied during the course of the experiment.

Measurement of nuclear shape

Following DAPI staining of nuclei, the nucleus boundaries in the fluores-

cence images were outlined with the Image J software, and nuclear shape

index (NSI) was calculated as NSI ¼ 4p � Area/(Perimeter)2. The NSI

assumes values between 1 (circular shape) and 0 (elongated, linear

morphology).

Nuclear protein extraction

MSCs were washed with cold phosphate-buffered saline (PBS), and lysed

with NucBuster Extraction Reagent 1 (EMD Biosciences, Gibbstown,

NJ). The lysates were vortexed for 15 s at high speed, incubated on ice

for 5 min, and then vortexed again for 15 s at high speed. The samples

were centrifuged at 16,000� g for 5 min at 4�C, and the supernatant (cyto-plasmic fraction) was removed. The pellets were washed with cold PBS,

and the pellet was resuspended in NucBuster Extraction Reagent 2. After

vortexing and centrifugation, the supernatant was collected as nuclear

extract. Protein concentration was determined by DC protein assay

(BioRad, Hercules, CA).

HDAC activity assay

HDAC activity was measured with a fluorometric HDAC assay kit (ab1438,

Abcam, Cambridge, UK). Nuclear extract was equilibrated with ddH2O to

85ml, resuspended in 10 ml 10X HDAC assay buffer, and transferred to

96-well plates. HDAC substrate, Boc-Lys(Ac)-AMC, was added and incu-

bated for 1 h at 37�C. Then, 10 ml lysine developer was added, and the

suspension was mixed well and incubated for 30 min. Relative HDAC

activity was determined in a fluorescence plate reader with excitation at

350–380 nm and emission at 440–460 nm and expressed as the relative fluo-

rescence units/mg protein sample.

Immunofluorescent staining and analysis

Immunostaining and confocal microscopy were used to visualize the struc-

ture of cytoskeleton and AcH3 in MSCs. MSCs were fixed with 4% para-

formaldehyde, permeabilized with 0.5% Triton X-100, and blocked with

1% bovine serum albumin. For cytoskeletal staining, samples were incu-

bated in rhodamine-conjugated phalloidin for 30 min to stain all filamen-

tous actin. For AcH3 staining, samples were incubated in anti-AcH3

primary antibody (Millipore, Billerica, MA) for 2 h, followed by incubation

with FITC-conjugated antirabbit IgG secondary antibody (Jackson Immu-

noResearch, West Grove, PA) for 1 h.

We used a Leica TCL SL confocal microscopy system, including He/Ne

laser sources and a Leica DM IRBmicroscope, to collect multiple Z-section

images (sections 0.3–0.5 mm thick over a range of 6 mm) for a given spec-

imen. These sections were subsequently projected to a single plane to create

an overall image of the specimen. All images in a given group were

collected with the same hardware and software settings.

Biophysical Journal 100(8) 1902–1909

1904 Li et al.

To assess the fluorescence intensity of AcH3 staining, nuclear fluores-

cence intensity was measured as the average of all pixel values within

the projected area of a nucleus. All image processing was performed using

Image J software.

Measurement of nuclear shape change inresponse to mechanical strain

MSCs were seeded on the PDMS elastic membranes with parallel micro-

grooves. After 24 h of culture, cells were loaded with DAPI (25 mg/ml)

in culture medium for 15 min. The stretch chambers were placed on the

stage of an Olympus upright microscope equipped with a 40� water-

immersion objective. Before imaging, cells were washed with Ringer’s

solution containing 155 mM NaCl, 4.5 mM KCl, 2 mM CaCl2, 1 mM

MgCl2, 10 mM D-glucose, and 5 mM Na-HEPES (pH 7.4). The PDMS

membranes with MSCs were subjected to 20% uniaxial static compres-

sion/stretch, and the nuclear shape was recorded by fluorescence micros-

copy before and after mechanical loading, followed by NSI calculation.

Knocking down lamin expression by using siRNA

The siRNA for lamin A/C was obtained from Dharmacon (Lafayette, CO).

A nonspecific control-pool siRNA (Dharmacon) was used as a negative

control for siRNA transfection. MSCs were transfected with siRNA

(100 nM for a culture area 10 cm2) using LipofectAMINE 2000 (Invitrogen,

Carlsbad, CA). The transfection efficiency of siRNAwas previously shown

to be >90% (16), and the siRNA exhibited maximum gene-knockdown

effectiveness at day 1–3 posttransfection (data not shown).

A B

Actin

10- m microgroovesNo-pattern

RNA isolation and quantitative polymerase chainreaction (qPCR)

Cells were lysed with RNA STAT-60 reagent (Tel-Test, Friendswood, TX).

RNAwas extracted by using chloroform and phenol extractions and precip-

itated by isopropanol, and the resulting RNA pellet was washed with 75%

ethanol. cDNA was synthesized by using two-step reverse transcription

(RT) with the ThermoScript RT-PCR system (Invitrogen), followed by

qPCR using SYBR green reagent and the ABI Prism 7000 Sequence Detec-

tion System (Life Tech, Carlsbad, CA). Primers for the genes of interest

were all designed by using the ABI Prism Primer Express software v.2.0

(Life Tech). The gene expression of each sample was normalized to the

level of 18S ribosomal RNA of the same sample.

50 m

le

i

C D

Statistical analysis

Mean and standard deviation (SD) values were calculated for each group of

data. The Student’s t-test was used to compare data between two experi-

mental groups. For comparisons among more than two groups, ANOVA

and a Holm’s t-test were used. Data was extracted from at least three exper-

iments. The asterisk indicates a statistically significant difference (p< 0.05)

between specified groups.

Nu

c

FIGURE 1 Immunofluorescent images showing MSCs on unpatterned

and micropatterned (10-mm-wide grooves) PDMS membranes after 24-h

culture. (A and B) Phalloidin staining of actin filaments on unpatterned

(A) and micropatterned (B) surfaces. (C and D) ToPro nuclear staining.

Arrows indicate the direction of microgrooves.

RESULTS

Parallel microgrooves modulated cellmorphology and nuclear shape

To determine the effects of micropatterned grooves onMSCs, cells were cultured on PDMS membranes with orwithout microgrooves for 1 day. Confocal microscopy ofactin filaments showed that MSCs spread out and had

Biophysical Journal 100(8) 1902–1909

random orientation on unpatterned elastic substrates(Fig. 1 A). In contrast, on micropatterned PDMSmembranes, MSCs demonstrated an elongated morphology,and the actin filaments aligned uniformly in the direction ofthe microgrooves (Fig. 1 B). However, the overall intensityand structure of the actin filaments did not show significantdifferences between patterned and unpatterned membranes.It is interesting to note that the elongated cell morphologywas accompanied by a more elongated nuclear shape anda partial alignment of the nucleus (Fig. 1, C and D). Quan-titative analysis showed that grooves 10 mm wide resulted insignificantly lower NSI values compared to the unpatternedsurface (Fig. 2 A), suggesting that the change in the cellshape and cytoskeleton could significantly affect the nuclearshape.

Culturing MSCs on micropatterned surfacesinduced alterations in HDAC activity and histoneacetylation level

To investigate whether the change in nuclear shape wasaccompanied by the remodeling of chromosome structure,we compared HDAC activity in MSCs on unpatterned andmicropatterned surfaces. Quantitative analysis indicatedthat HDAC activity in MSCs grown on the micropatternedsurfaces was much lower than that in MSCs cultured onthe unpatterned surfaces (Fig. 2 B). On the other hand,when we treated cells with CHAHA (1.5–15 mM), a chemi-cal inhibitor of HDAC that increased histone acetylation, we

0 6

0.7

0.8

0.9

1

NS

I

*

*

0.5

0.6

10- mNo-pattern

B

A

HD

AC

Activity

10

20

30

40

50

60

No-pattern

H

0

10

10- m

FIGURE 2 Effects of micropatterning on nuclear shape and HDAC

activity of MSCs. MSCs were cultured on unpatterned and micropatterned

(10-mm-wide grooves) surfaces for 24 h. (A) Nuclear shape was measured,

and NSI was calculated. (B) Nuclear fraction of MSCs was used for HDAC

activity assay. Bars represent mean 5 SD. Asterisk indicates the statisti-

cally significant difference (p < 0.05).

No-pattern

A

B

C

FIGURE 3 Analysis of histone acetylation in MSCs on unpatterned and

Biophysical Regulation of HDAC 1905

did not observe a significant change in nuclear shape (datanot shown).

We then determined whether the alteration of HDACactivity by the micropatterned surface resulted in changesin histone acetylation. Indeed, we showed that MSCs grownon the micropatterned surfaces had a significant increase inhistone 3 acetylation (Fig. 3), which was consistent with thedecrease of HDAC activity.

micropatterned PDMS membranes. (A and B) Immunofluorescence images

of AcH3 in MSCs on unpatterned (A) and micropatterned surfaces (B).

(C) Quantitative measurement of AcH3 levels (relative fluorescence inten-

sity) in MSCs cultured on unpatterned and micropatterned surfaces. Arrow

indicates the direction of microgrooves. Bars represent mean 5 SD.

Asterisk indicates the statistically significant difference (p < 0.05) between

specified groups.

Regulation of HDAC activity by anisotropicmechanical loading

We first investigated whether mechanical loading modulatedHDAC activity in MSCs on unpatterned surfaces. As shownin Fig. 4 A, mechanical compression did not significantlyaffect HDAC activity when cells were randomly oriented.To further determine whether anisotropic mechanicalloading modulated HDAC activity in MSCs, we measuredHDAC activity of aligned MSCs in response to differenttypes and directions of mechanical load. MSCs werecultured on micropatterned membranes, and a one-stepuniaxial strain of 20% compression or stretch was appliedto MSCs in the direction parallel or perpendicular to the

axis of the microgrooves, i.e., the cell alignment direction.With 20% compression, HDAC activity was significantlydownregulated when the compression or stretch was perpen-dicular to the axis of cell alignment (Fig. 4). However,compression or stretch in the direction parallel to the axisof cell alignment did not significantly affect HDAC activity.These results suggest that the effect of mechanical strain onHDAC activity was dependent on the direction of strain.

Biophysical Journal 100(8) 1902–1909

0.6

0.8

1.0

1.2

1.4

AC

Ac

tiv

ity

A

B

0.0

0.2

0.4

HD

A

Compression

(No pattern)

Control

DA

C A

ctiv

ity

0 4

0.6

0.8

1.0

1.2

*

C

HD

0.0

0.2

0.4

Parallel

compression

Perpendicular

compression

AC

Ac

tiv

ity

0.6

0.8

1.0

1.2

*

HD

A

0.0

0.2

0.4

Parallel

stretch

Perpendicular

stretch

FIGURE 4 Differential alterations of HDAC activity in the nuclei of

MSCs by static uniaxial mechanical strain. Static uniaxial strain (20%

compression or 20% stretch) was applied to unpatterned or micropatterned

PDMS membranes (either in parallel or perpendicular to microgrooves) for

2 h. Control samples were not subjected to mechanical strain. Nuclear

protein of MSCs was extracted for HDAC activity assay. (A) HDAC activity

of MSCs on unpatterned surfaces in response to compression. (B) The ratio

of HDAC activity (compression/control) of MSCs on micropatterned

membranes in response to compression. (C) The ratio of HDAC activity

(stretch/control) of MSCs on micropatterned membranes in response to

stretch. Bars represent mean 5 SD. Asterisk indicates the statistically

Biophysical Journal 100(8) 1902–1909

1906 Li et al.

Regulation of histone acetylation by anisotropicmechanical loading

To confirm the effects of perpendicular mechanical strain onhistone acetylation, we measured the acetylation of histoneH3 as a representative. As shown in Fig. 5, indeed thecompression or stretch perpendicular to the cell alignmentsignificantly increased histone H3 acetylation, consistentwith the results on HDAC activity.

Effects of anisotropic mechanical loadingon nuclear deformation

We further determined whether anisotropic mechanicalstrain induced changes in nuclear shape. Fig. 6 showsnuclear deformation in MSCs before and after 20% staticcompression or stretch either parallel or perpendicular tothe axis of the microgrooves. It was interesting that onlycompression and stretch in the perpendicular direction hadsignificant effects on nuclear shape, although the changesof NSI (~0.02) induced by mechanical strain were muchless than the changes of NSI induced by microgrooves(~0.1). Compression in the perpendicular direction madethe cell nuclei more elongated, whereas stretch in theperpendicular direction made cell nuclei less elongated,suggesting that nuclear shape change was more sensitiveto mechanical strain in the direction perpendicular to cellalignment. Since compression and stretch had oppositeeffects on nuclear shape, and the change of NSI was small,the effect of mechanical strain on HDAC activity may not beregulated by a change in cell shape.

Role of lamin A/C in mechanical strain-regulatedHDAC activity

To determine whether nuclear matrix mediated mechano-transduction, we knocked down lamin A/C, a major struc-tural component of nuclear matrix. Lamin A/C siRNAsuppressed the expression of lamin A/C by 50% (Fig. 7A). Knocking down lamin A/C had no effect on nuclearshape on either unpatterned or micropatterned surfaces,and NSI was still significantly lower on micropatternedsurfaces (Fig. 7 B). Since mechanical compression inducedmore change in HDAC activity than stretch (Fig. 4), we usedcompression as a representative model to determine whetherlamin A/C mediated strain-modulated HDAC activity. ForMSCs treated with control siRNA, mechanical compressionsuppressed HDAC activity (Fig. 7 C), as shown previously(Fig. 4); however, knocking down lamin A/C abolishedthis effect, suggesting that lamin A/C played an importantrole in mechanotransduction leading to HDAC activitychange.

significant difference (p < 0.05) compared to the respective control group.

Log-transformed t-test was used to compare the samples with respective

controls. Dotted lines indicate the relative level of controls.

A

Control Compression

Stretch

B

CA

cH

3

D

8

10

12 **

Level o

f A

0

2

4

6

8

FIGURE 5 Effects of perpendicular stretch and compression on histone

acetylation in MSCs on elastic membranes. Uniaxial stretch or compression

(20%) was applied to the micropatterned membranes with MSCs for 2 h in

the direction perpendicular to the microgrooves. (A–C) Immunofluores-

cence images of AcH3 in MSCs without strain (A), subjected to compres-

sion (B), or subjected to stretch. (D) Measurement of AcH3 levels in

MSCs. Arrows indicate the direction of microgrooves. Bars represent

mean 5 SD. Asterisk indicates the statistically significant difference

(p < 0.05) between specified groups.

1

pre

A

NS

I

0.6

0.7

0.8

0.9post

*

0.5

Parallel

compression

Perpendicular

compression

0.8

0.9

1pre

post

*

NS

I

B

0.5

0.6

0.7

Parallel

stretch

Perpendicular

stretch

FIGURE 6 Effects of static mechanical load on nuclear shape. MSCs

were cultured on micropatterned membranes, stained with DAPI, and

subjected to 20% static uniaxial compression (A) or stretch (B) in a direction

parallel or perpendicular to the microgrooves. The images of nuclei were

collected from 10 fields of each sample before and after stretch or compres-

sion. Nuclear shape was measured, and NSI was calculated. Bars represent

mean 5 SD. Asterisk indicates the statistically significant difference

(p < 0.05).

Biophysical Regulation of HDAC 1907

DISCUSSION

Local microenvironmental factors have profound effects oncell shape, cytoskeletal organization, nuclear matrix organi-zation, chromatin structure, and gene expression. In thisstudy, micropatterning and mechanical loading were usedto examine how cell adhesion substrate and anisotropicmechanical strain regulated nuclear shape, HDAC activity,and histone acetylation. Here, we showed, for the firsttime that we know of, that biophysical cues provided bythe substrates, either microtopographic patterning or aniso-tropic mechanical strain, could modulate HDAC activityand histone acetylation. These findings reveal a new mech-anism, to our knowledge, by which biophysical factors regu-late gene expression. It is generally accepted that ECM isdynamically coupled to the nucleus through the cytoskel-eton and the nuclear matrix (17). For example, there isevidence that the nuclear envelope plays an important rolein mechanotransduction (18–20). However, how thesemechanical couplings regulate gene expression is not clear.Our results suggest that mechanical signals can be convertedinto chemical signaling such as HDAC activity and histoneacetylation in a nuclear-matrix-dependent manner, thus

modulating the structure of chromosomes and the patternof gene expression. Besides histone acetylation, other modi-fications of histones, such as phosphorylation, methylation,and ribosylation, could also be regulated by biophysicalfactors, which remains to be determined in future studies.

An interesting finding is that the nuclear shape could berelated to the HDAC activity in the nucleus. MSCs onmicrogrooves had elongated nuclei, a decrease in HDACactivity, and an increase in histone acetylation. However,mechanical compression in the perpendicular directioninduced a more elongated nuclear shape and a decrease ofHDAC activity, whereas mechanical stretch in the perpen-dicular direction resulted in a less elongated nuclear shapeand a decrease of HDAC activity. One explanation is thatthe effects of micropatterning and mechanical loading

Biophysical Journal 100(8) 1902–1909

A 1

Lam

in

A

/C

0

0.5

B Control siRNA

NS

I

0.7

0.8

0.9

1.0

Lamin siRNA

0.5

0.6

No pattern 10- m

C

2000

3000

4000

DA

C A

ctivity

Control siRNA

Lamin siRNA

*

0

1000HD

FIGURE 7 Role of lamin A/C in mechanotransduction. (A) Knocking

down lamins A and C by using siRNA. After 2 days, gene expression

was quantified by qPCR. (B) Effect of knocking down lamin A/C (day 2)

on nuclear morphology on unpatterned or patterned surfaces. (C) MSCs

transfected with control siRNA or lamin A/C siRNA were subjected to

Biophysical Journal 100(8) 1902–1909

1908 Li et al.

involve different mechanisms. The effect of elongatednuclear shape could be related to the difference in nuclearpores (17) and spatial distribution of HDAC and HDACregulators, whereas the effect of mechanical loading maybe due to the change in nuclear membrane and nuclearmatrix. For example, the change in nuclear shape may affectthe rate of nucleocytoplasmic transport (21), which in turnregulates the shuttling of HDACs and their regulators.Biochemically, the shuttling of HDAC is regulated by phos-phorylation, which changes their binding affinity to chap-erone proteins that go in or out of the nucleus (22). Thesepossibilities await further investigation.

Another important finding is that mechanosensing andHDAC regulation are anisotropic. HDAC activity wasdecreased by compression or stretch in the perpendiculardirection of cell alignment, but was not affected bycompression or stretch in the longitudinal direction ofaligned cells. The mechanism of anisotropic mechanosens-ing is not clear. It is possible that aligned cells have aniso-tropic assembly and organization of the cytoskeleton andintracellular organelles, which result in different distributionof stress and strain in the cells.

Our results also indicate that nuclear matrix protein laminplays an important role in the downregulation of HDACactivity in response to mechanical strain. Lamins A and Care required for the maintenance of nuclear mechanics, aslamin-A/C-deficient cells have severely reduced nuclearstiffness and decreased cell viability under strain (15). It isinteresting to note that partially knocking down lamin A/Cdid not significantly affect nuclear shape and cell viability,which provides an experimental system to study the roleof lamin in mechanotransduction. How lamin regulatesHDAC activity is not known. It can be related to nuclearmechanics and/or protein transport across the nuclear enve-lope, which remains to be determined in future studies.

The regulation of nuclear shape and HDAC activity bymicropatterning and mechanical strain may be cell-typedependent, and can be related to the size, cytoskeletal struc-ture, and mechanical property of the cells. For example,vascular smooth muscle cells on the same type of micropat-terned surfaces did not show significant change in HDACactivity compared to cells on unpatterned surfaces. Mesen-chymal cells, epithelial cells, and neuronal cells are verydifferent in subcellular structure, and their responses to mi-cropatterning and mechanical strain may be quite different.

Uniaxial strain is relevant to mechanical loading in manytypes of tissues in vivo, e.g., heart, vascular wall, muscle,cartilage, tendon, and ligament. Therefore, the resultsfrom this in vitro cell culture system may have significantimplications. Previous studies have demonstrated the aniso-tropic responses of cells to cyclic mechanical strain (6,23–25), including gene expression, proliferation, cytoskeleton

20% compression for 2 h, followed by HDAC activity assay. Asterisk indi-

cates the statistically significant difference (p < 0.05).

Biophysical Regulation of HDAC 1909

rearrangement, and phenotypic change. Since static andcyclic mechanical strain may have different effects, itremains to be determined whether cyclic strain regulatesHDAC activity and whether HDAC activity mediates cyclicstrain-induced gene expression. Since ECM and mechanicalloading influence cell morphology and gene expression ina wide variety of tissues, it is likely that the change in globalhistone acetylation is a common mechanism in the regula-tion of gene expression by these biophysical factors.Because HDAC activity and histone acetylation regulatethe expression of many genes involved in cell proliferation,differentiation, and migration, the understanding ofbiophysical regulation of HDAC activity will provide mech-anistic insights into cell functions during tissue regenera-tion. Elucidating the anisotropic mechanosensing andchromatin remodeling of cells will not only advance ourunderstanding of the roles of mechanical strain in tissueremodeling but also provide a rational basis to design biore-actors for tissue engineering that can utilize appropriatemechanical stimulation.

We thank Junyu Mai, Qian Chen, Randall R. R. Janairo, and Kristin A.Ger-

hold for their technical assistance.

This research was supported in part by grants from the National Institutes of

Health (HL078534 and HL083900, to S.L.), the National Science Founda-

tion (CMS-0528506, to K.P.S.), and the Chinese National Science Founda-

tion (the 111 Program and 973 project 2005CB522703, to L.Y.).

REFERENCES

1. Jiang, Y., B. N. Jahagirdar, ., C. M. Verfaillie. 2002. Pluripotency ofmesenchymal stem cells derived from adult marrow. Nature.418:41–49.

2. Caplan, A. I., and S. P. Bruder. 2001. Mesenchymal stem cells: buildingblocks for molecular medicine in the 21st century. Trends Mol. Med.7:259–264.

3. Prockop, D. J. 1997. Marrow stromal cells as stem cells for nonhema-topoietic tissues. Science. 276:71–74.

4. Pittenger, M. F., J. D. Mosca, and K. R. McIntosh. 2000. Humanmesenchymal stem cells: progenitor cells for cartilage, bone, fat andstroma. Curr. Top. Microbiol. Immunol. 251:3–11.

5. Huang, N. F., and S. Li. 2008. Mesenchymal stem cells for vascularregeneration. Regen. Med. 3:877–892.

6. Kurpinski, K., J. Chu,., S. Li. 2006. Anisotropic mechanosensing bymesenchymal stem cells. Proc. Natl. Acad. Sci. USA. 103:16095–16100.

7. Fullgrabe, J., N. Hajji, and B. Joseph. 2010. Cracking the death code:apoptosis-related histone modifications. Cell Death Differ. 17:1238–1243.

8. Kouzarides, T. 2007. Chromatin modifications and their function. Cell.128:693–705.

9. Haberland, M., R. L. Montgomery, and E. N. Olson. 2009. The manyroles of histone deacetylases in development and physiology: implica-tions for disease and therapy. Nat. Rev. Genet. 10:32–42.

10. Stewart, C. L., K. J. Roux, and B. Burke. 2007. Blurring the boundary:the nuclear envelope extends its reach. Science. 318:1408–1412.

11. Dahl, K. N., A. J. Ribeiro, and J. Lammerding. 2008. Nuclear shape,mechanics, and mechanotransduction. Circ. Res. 102:1307–1318.

12. Galiova, G., E. Bartova, ., S. Kozubek. 2008. Chromatin changesinduced by lamin A/C deficiency and the histone deacetylase inhibitortrichostatin A. Eur. J. Cell Biol. 87:291–303.

13. Thakar, R. G., Q. Cheng, ., S. Li. 2009. Cell-shape regulation ofsmooth muscle cell proliferation. Biophys. J. 96:3423–3432.

14. Le Beyec, J., R. Xu,., M. J. Bissell. 2007. Cell shape regulates globalhistone acetylation in human mammary epithelial cells. Exp. Cell Res.313:3066–3075.

15. Lammerding, J., P. C. Schulze, ., R. T. Lee. 2004. Lamin A/C defi-ciency causes defective nuclear mechanics and mechanotransduction.J. Clin. Invest. 113:370–378.

16. Wang, D., J. S. Park, ., S. Li. 2004. Proteomic profiling of bonemarrow mesenchymal stem cells upon transforming growth factor b1stimulation. J. Biol. Chem. 279:43725–43734.

17. Wang, N., J. D. Tytell, and D. E. Ingber. 2009. Mechanotransduction ata distance: mechanically coupling the extracellular matrix with thenucleus. Nat. Rev. Mol. Cell Biol. 10:75–82.

18. Lammerding, J., J. Hsiao,., R. T. Lee. 2005. Abnormal nuclear shapeand impaired mechanotransduction in emerin-deficient cells. J. CellBiol. 170:781–791.

19. Webster, M., K. L. Witkin, and O. Cohen-Fix. 2009. Sizing up thenucleus: nuclear shape, size and nuclear-envelope assembly. J. CellSci. 122:1477–1486.

20. Khatau, S. B., C. M. Hale, ., D. Wirtz. 2009. A perinuclear actin capregulates nuclear shape. Proc. Natl. Acad. Sci. USA. 106:19017–19022.

21. Feldherr, C. M., and D. Akin. 1990. The permeability of the nuclearenvelope in dividing and nondividing cell cultures. J. Cell Biol.111:1–8.

22. Legube, G., and D. Trouche. 2003. Regulating histone acetyltrans-ferases and deacetylases. EMBO Rep. 4:944–947.

23. Wang, J. H., E. S. Grood,., R. Wenstrup. 2000. Alignment and prolif-eration of MC3T3-E1 osteoblasts in microgrooved silicone substratasubjected to cyclic stretching. J. Biomech. 33:729–735.

24. Gopalan, S. M., C. Flaim, ., A. D. McCulloch. 2003. Anisotropicstretch-induced hypertrophy in neonatal ventricular myocytes micro-patterned on deformable elastomers. Biotechnol. Bioeng. 81:578–587.

25. Houtchens, G. R., M. D. Foster,., J. Y. Wong. 2008. Combined effectsof microtopography and cyclic strain on vascular smooth muscle cellorientation. J. Biomech. 41:762–769.

Biophysical Journal 100(8) 1902–1909


Recommended