Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Jyothish B. Pillai, Madhu Gupta, Senthilkumar B.

Rajamohan, Roberto Lang, Jai


Raman and Mahesh P. Gupta
Am J Physiol Heart Circ Physiol 291:1545-1553, 2006. First published Apr 21, 2006;
doi:10.1152/ajpheart.01124.2005

You might find this additional information useful...

This article cites 35 articles, 21 of which you can access free at:
http://ajpheart.physiology.org/cgi/content/full/291/4/H1545#BIBL

This article has been cited by 3 other HighWire hosted articles:


PARP-1 suppresses adiponectin expression through poly(ADP-ribosyl)ation of
PPAR{gamma} in cardiac fibroblasts
D. Huang, C. Yang, Y. Wang, Y. Liao and K. Huang
Cardiovasc Res, January 1, 2009; 81 (1): 98-107.
[Abstract] [Full Text] [PDF]

Metallothionein Suppresses Angiotensin II-Induced Nicotinamide Adenine Dinucleotide


Phosphate Oxidase Activation, Nitrosative Stress, Apoptosis, and Pathological Remodeling
in the Diabetic Heart
G. Zhou, X. Li, D. W. Hein, X. Xiang, J. P. Marshall, S. D. Prabhu and L. Cai

Downloaded from ajpheart.physiology.org on March 16, 2009


J. Am. Coll. Cardiol., August 19, 2008; 52 (8): 655-666.
[Abstract] [Full Text] [PDF]

Mice expressing ACE only in the heart show that increased cardiac angiotensin II is not
associated with cardiac hypertrophy
H. D. Xiao, S. Fuchs, E. A. Bernstein, P. Li, D. J. Campbell and K. E. Bernstein
Am J Physiol Heart Circ Physiol, February 1, 2008; 294 (2): H659-H667.
[Abstract] [Full Text] [PDF]

Updated information and services including high-resolution figures, can be found at:
http://ajpheart.physiology.org/cgi/content/full/291/4/H1545

Additional material and information about AJP - Heart and Circulatory Physiology can be found at:
http://www.the-aps.org/publications/ajpheart

This information is current as of March 16, 2009 .

AJP - Heart and Circulatory Physiology publishes original investigations on the physiology of the heart, blood vessels, and
lymphatics, including experimental and theoretical studies of cardiovascular function at all levels of organization ranging from the
intact animal to the cellular, subcellular, and molecular levels. It is published 12 times a year (monthly) by the American
Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2005 by the American Physiological Society.
ISSN: 0363-6135, ESSN: 1522-1539. Visit our website at http://www.the-aps.org/.
Am J Physiol Heart Circ Physiol 291: H1545–H1553, 2006.
First published April 21, 2006; doi:10.1152/ajpheart.01124.2005.

Poly(ADP-ribose) polymerase-1-deficient mice are protected from angiotensin


II-induced cardiac hypertrophy
Jyothish B. Pillai,1 Madhu Gupta,4 Senthilkumar B. Rajamohan,1
Roberto Lang,2 Jai Raman,1 and Mahesh P. Gupta1,3
Departments of 1Surgery and 2Cardiology, and 3Committee on Molecular Medicine and Pathology, The University
of Chicago, Chicago, Illinois; and 4The Heart Institute of Children, Hope Children Hospital, Oak Lawn, Illinois
Submitted 24 October 2005; accepted in final form 12 April 2006

Pillai, Jyothish B., Madhu Gupta, Senthilkumar B. Rajamo- established as a most potent stimulator of cardiac hypertro-
han, Roberto Lang, Jai Raman, and Mahesh P. Gupta. Poly- phy. Multiple signaling pathways that regulate angiotensin
(ADP-ribose) polymerase-1-deficient mice are protected from an- II-mediated cardiac hypertrophy response have been also
giotensin II-induced cardiac hypertrophy. Am J Physiol Heart Circ identified; these include activation of PKC, MAPKs, and the
Physiol 291: H1545–H1553, 2006. First published April 21, 2006; production of reactive oxygen and nitrogen species (30).
doi:10.1152/ajpheart.01124.2005.—Poly(ADP-ribose) polymer-
However, the downstream targets and nuclear integrators of
ase-1 (PARP), a chromatin-bound enzyme, is activated by cell
angiotensin II-mediated cardiac growth response are not yet

Downloaded from ajpheart.physiology.org on March 16, 2009


oxidative stress. Because oxidative stress is also considered a main
component of angiotensin II-mediated cell signaling, it was pos- understood.
tulated that PARP could be a downstream target of angiotensin Poly(ADP-ribose) polymerase-1 (PARP) is a (113 kDa)
II-induced signaling leading to cardiac hypertrophy. To determine multifunctional chromatin bound enzyme of the PARP family
a role of PARP in angiotensin II-induced hypertrophy, we infused of proteins. PARP catalyzes the transfer of multiple units of the
angiotensin II into wild-type (PARP⫹/⫹) and PARP-deficient mice. ADP-ribose moiety from NAD to the target proteins, a process
Angiotensin II infusion significantly increased heart weight-to- called poly-ADP-ribosylation (4, 18). PARP is activated by
tibia length ratio, myocyte cross-sectional area, and interstitial DNA single-strand break as well as by production of oxygen
fibrosis in PARP⫹/⫹ but not in PARP⫺/⫺ mice. To confirm these and nitrogen-derived free radicals and nuclear accumulation of
results, we analyzed the effect of angiotensin II in primary cultures Ca2⫹ and Mg2⫹ ions (4, 17). In physiological conditions, mild
of cardiomyocytes. When compared with PARP⫺/⫺ cardiomyo- PARP activation regulates many cellular processes, including
cytes, angiotensin II (1 ␮M) treatment significantly increased DNA repair, gene transcription, cell-cycle progression, cy-
protein synthesis in PARP⫹/⫹ myocytes, as measured by 3H- toskeleton organization, cell survival, and chromatin remodel-
leucine incorporation into total cell protein. Angiotensin II-medi-
ing (4, 18, 20). However, overactivation of PARP threatens cell
ated hypertrophy of myocytes was accompanied with increased
poly-ADP-ribosylation of nuclear proteins and depletion of cellu-
survival as it consumes cellular NAD content to add extended
lar NAD content. When cells were treated with cell death-inducing chains of ADP-ribose moieties on the target proteins (4, 18).
doses of angiotensin II (10 –20 ␮M), robust myocyte cell death was Because NAD is essential for many crucial cell processes,
observed in PARP⫹/⫹ but not in PARP⫺/⫺ myocytes. This type of including energy metabolism, intracellular Ca2⫹ regulation,
cell death was blocked by repletion of cellular NAD levels as well and the activity of class-III histone deacetylases (also called
as by activation of the longevity factor Sir2␣ deacetylase, indicat- sirtuins or Sir2), depletion of NAD rapidly leads to functional
ing that PARP induction and subsequent depletion of NAD levels deficit and eventually to cell death (7a, 23). Overactivation of
are the sequence of events causing angiotensin II-mediated cardi- PARP was implicated in various pathological conditions asso-
omyocyte cell death. In conclusion, these results demonstrate that ciated with oxidative cell stress, including inflammation, dia-
PARP is a nuclear integrator of angiotensin II-mediated cell betes mellitus, circulatory shock, stroke, and reperfusion injury
signaling contributing to cardiac hypertrophy and suggest that this of many organs, including heart, brain, and kidney (reviewed
could be a novel therapeutic target for the management of heart in Refs. 18 and 21). Data obtained from using PARP inhibitors
failure. have also documented a role of PARP activation in the devel-
heart failure; oxidative-stress signaling opment and progression of pressure-overload cardiac hypertro-
phy and heart failure (32).
Here we report that PARP is activated during angiotensin
IN SETTINGS OF sustained hemodynamic overload, myocardium II-induced cardiomyocyte hypertrophy, and mice deficient in
undergoes a growth response, termed as cardiac hypertrophy. the PARP gene are protected from angiotensin II-mediated
This process is characterized by an increase in myocyte cell hypertrophy. We also show that NAD depletion, which results
size and alterations in expression of contractile and Ca2⫹- from PARP overactivation, contributes to angiotensin II-medi-
handling proteins. Cardiac hypertrophy initially develops as an ated cardiac myocyte cell death. This type of cell death was
adaptive response; however, if remained unchecked, this even- prevented by NAD repletion and activation of the longevity
tually progresses to ventricular dilation and pump dysfunction factor Sir2␣ (3). These results demonstrate that PARP is a
(12). At the biochemical level, many hypertrophy agonists downstream nuclear target of angiotensin II-induced signaling
have been identified; among them, angiotensin II has been pathway, contributing to cardiac hypertrophy and failure.

Address for reprint requests and other correspondence: M. P. Gupta, Dept. The costs of publication of this article were defrayed in part by the payment
of Surgery, MC 5040, Univ. of Chicago, 5841 S. Maryland Ave., Chicago, IL of page charges. The article must therefore be hereby marked “advertisement”
60637 (e-mail: mgupta@surgery.bsd.uchicago.edu). in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

http://www.ajpheart.org 0363-6135/06 $8.00 Copyright © 2006 the American Physiological Society H1545
H1546 HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II

METHODS 33342 and propidium iodide dyes for 5 min. Cells that detached from
the plate were collected and included in the analysis. Cells were
Mice. PARP ⫺/⫺
mice (C57BL/6 ⫻ 129 background) were pur- washed twice and resuspended in PBS and analyzed by fluorescence-
chased from Jackson Laboratories. Cardiac hypertrophy and failure activated cell sorting (FACS) analysis.
was induced in adult (8 wk old) wild-type (PARP⫹/⫹) and PARP⫺/⫺ [3H]Leucine incorporation. Immediately after treatment of cardi-
mice of similar genetic background, by chronic infusion of angioten- omyocytes with angiotensin II, cells were incubated with [3H]leucine
sin II (Sigma) at a subpressure dose of 0.3 mg 䡠 kg⫺1 䡠 day⫺1 for 14 (1.0 mCi/ml, 167 Ci/mmol Specific activity, Amersham Biosciences)
days by using Alzet osmotic minipumps (Alzet model 1002). Pumps in leucine-free MEM medium (Invitrogen) for 72 h. To precipitate
were removed before echocardiographic measurements. All experi- proteins, cells were washed with PBS and then incubated in 10%
mental protocols were approved by the University of Chicago’s trichloroacetic acid. The resultant pellet was solublized in NaOH
Animal Care and Use Committee. (0.25 N), and the lysate was diluted with one-sixth volume of
Assessment of cardiac hypertrophy. Mice were euthanized, and scintillation fluid and counted in a scintillation counter. Values were
body weight, heart weight, and tibia length were recorded. Some normalized with DNA content, which was measured by using
hearts were fixed and cryosectioned (5 ␮m) for morphometric anal- Quant-iT picogreen dsDNA assay kit (Invitrogen).
yses. To determine myofibrillar organization, sections were immuno- NAD estimation. The concentration of NAD was measured as
stained with anti-␣-actinin primary antibody (A-7811) and anti-mouse described previously (35) with slight modifications. The samples
IgG FITC (F-2012) as secondary antibody (Sigma). For measurement (myocyte cultures or 50 g frozen crushed tissue) were resuspended in
of interstitial fibrosis, sections were stained with Masson trichrome 200 ␮l of 0.5 M perchloric acid. Cell extracts were neutralized with
stain. Collagen content was quantified by assessing the blue pixel equal volume of 1 M KOH and 0.33 M KH2PO4/K2HPO4 (pH 7.5),
content in a computer-assisted image analysis program. Myocyte cell centrifuged to remove the KClO4 precipitate, and the supernatant was
size (⬎100 cells/section) was measured from transverse section collected. To 200 ␮l of NAD reaction mixture (600 mM ethanol, 0.5

Downloaded from ajpheart.physiology.org on March 16, 2009


stained with wheat germ agglutinin (100 ␮g/ml) coupled to tetra- mM 3-[4,5 dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide, 2
methylrhodamine isothiocyanate (Sigma) by using a digital image mM phenazine ethosulfate, 5 mM EDTA, 1 mg/ml BSA, and 120 mM
analyzer program. bicine at pH 7.8), 50 ␮l of supernatant or NAD standard were added
Cell culture and transfection. Primary cultures of cardiac myocytes and incubated for 5 min at 37°C. Twenty-five microliters of alcohol
were obtained from 1-day-old mouse neonatal pups according to the dehydrogenase (0.5 mg/ml in 100 mM bicine at pH 7.8) were added
procedure described previously (8). Briefly, hearts were quickly to the reaction mix and incubated for an additional 20 min at 37°C.
excised, atria were removed, and the ventricles were collected in The reaction was stopped by adding 250 ␮l of 12 mM iodoacetate, and
ice-cold Ca2⫹ and Mg2⫹-free HBSS, pH 7.4. Ventricles were minced the optical density was read at 570-nm wavelength. A standard curve
and digested in trypsin solution that was made in HBSS (0.1%) at was generated from known concentrations of NAD and used to
37°C for 8 min. Trypsin was then inactivated with an equal volume of calculate the concentration of NAD in each sample.
FBS, and cells were collected by centrifugation. The first digested PARP assay. PARP activity of cardiac myocyte cultures treated
cells were discarded. The remaining tissue was digested for four more with different hypertrophy agaonists was measured using Universal
cycles, and the cells that were collected from each cycle were colorimetric PARP assay kit (4672– 096K, Trevigen), according to the
suspended in MEM supplemented with 10% FBS and 5 mg/ml manufacturer’s protocol. Briefly, the PARP enzyme activity of each
penicillin-streptomycin (Invitrogen). Cell suspension was filtered and sample (cell lysate) was estimated based on the incorporation of
centrifuged, and cell pellet was suspended in MEM. Fibroblasts were biotinylated poly-ADP-ribose units onto histone proteins coated in a
removed by preplating twice for 45 min each, and myocytes were 96-well plate that was provided with the kit. The values were calcu-
further purified by Percoll density gradient. Finally, myocytes were lated from a standard curve generated by using known amounts of
collected by centrifugation, counted, and plated in laminin-coated PARP enzyme and normalized to the protein content.
culture dishes. Cells were grown in MEM (Invitrogen) for 48 h and
then treated and/or transfected by using TfX-TM-20 reagent (Pro- RESULTS
mega). Typically, 1 ⫻ 105 cells were transfected with 1.0 ␮g of
plasmid expressing NAD-biosynthetic enzyme nicotinamide phospho- PARP-deficient mice failed to produce angiotensin II-medi-
ribosyl-transferase (NAMPT) or Sir2␣ deacetylase. Resveratrol and ated hypertrophy response. To determine the role of PARP in
NAD were purchased from Sigma; small interfering RNA (siRNA)- angiotensin II-mediated cardiac hypertrophy, we chronically
mediated silencing of Sir2␣ was carried out by transient transfection infused angotensin II into control (PARP⫹/⫹) and PARP-
of pSUPER RNAi system (VEC-PBS-0003/0004) with the incorpo- deficient (PARP⫺/⫺) mice (C57BL/6 ⫻ 129 background).
ration of 20 nucleotides of Sir2␣ (gaagttgacctcctcattgt).
PARP knockout mice, in general, were smaller in size and had
Western and Northern blot analyses. Western blot analysis was
performed according to standard procedures using rabbit anti-Sir2␣ slightly higher heart weight-to-tibia length ratio and a little
antibody (Upstate) and anti-polyADP-ribose antibody (Alexis Bio- larger cross-sectional area of ventricular myocytes compared
chemicals). For RNA analysis, total heart RNA was analyzed by with age-matched control mice (Fig. 1, A and E). Angiotensin
Northern blot analysis using gene-specific probes as described previ- II infusion in control mice produced a marked increase in heart
ously (24). For quantification purposes, autoradiograms were scanned size, heart weight-to-tibia length ratio, and left ventricular mass
using Scion Image Windows analysis software based on NIH Image and wall thickness (Table 1). The left ventricular end-diastolic
for Macintosh by Wayne Rasband. Signal density was adjusted for and end-systolic dimensions were also significantly increased
background density of the blot and calculated as percent change from in control mice (Table 1), reflecting signs of ventricular dila-
vehicle-treated controls. tation. Angiotensin II-infused PARP⫺/⫺ mice did not produce
Measurement of cell death. Myocytes were stained with two DNA a significant amount of cardiac hypertrophy, and also no signs
binding dyes (Molecular Probes): Hoechst 33342, which is permeable
to cell membrane and stains both live and dead cells blue, and
of ventricular dilation were observed (Table 1). Consistent
propidium iodide, which is impermeable to intact cell membrane and with these observations, the cross-sectional area of ventricular
stains dead cells red. For cell death measurements, adherent cells and myocytes was significantly increased in control mice but not in
detached cells were processed separately. Briefly, myocytes were PARP-deficient mice, thus confirming impaired hypertrophy
trypsinized and collected by centrifugation. A single cell suspension response in PARP⫺/⫺ mice. Angiotensin II-mediated cardiac
of myocytes was prepared in PBS and stained with both Hoechst hypertrophy is known to be associated with typical induction of
AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org
HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II H1547

Downloaded from ajpheart.physiology.org on March 16, 2009


Fig. 1. Cardiac hypertrophy response of poly(ADP-ribose) polymerase (PARP⫹/⫹) and PARP⫺/⫺ mice to ANG II infusion. A: quantification of heart
weight-to-tibia length ratio (HW/TL) in vehicle and ANG II-treated mice (n ⫽ 12/group; *P ⬍ 0.01). B: percent change in HW/TL (*P ⬍ 0.001). C:
quantification of cardiac hypertrophy marker mRNA transcripts in PARP⫹/⫹ and PARP⫺/⫺ mice (n ⫽ 3 for each gene; *P ⬍ 0.001). D: representative heart
sections (⫻200) stained with wheat germ agglutinin coupled with tetramethylrhodamine isothiocynate to demarcate cell boundaries. E: quantification of
cardiomyocyte size based on cross-sectional area in vehicle and ANG II-treated mice (n ⫽ 4 in each group, with 5 sections from each heart; *P ⬍ 0.05). NS,
not significant. Values are means ⫾ SE.

the fetal gene program. We therefore measured mRNA levels mRNA was significantly reduced in control mice. The expres-
of hypertrophy marker genes in PARP⫹/⫹ and PARP⫺/⫺ mice sion profile of these genes, however, was not significantly
after infusion of angiotensin II for 2 wk. As shown in Fig. 1C, altered in PARP-deficient mice, thus indicating a loss of the
expression levels of atrial natriuretic factor and ␤-myosin typical angiotensin II-mediated hypertrophy response in
heavy chain (MHC) mRNA was increased, and ␣-MHC PARP⫺/⫺ mice.

Table 1. Echocardiographic parameters in PARP⫹/⫹ and PARP⫺/⫺ mice after ANG II infusion for 2 wk
PARP⫹/⫹ PARP⫺/⫺

Vehicle ANG II Vehicle ANG II

HR, beats/min 462⫾15.5 545⫾14.6 353⫾12.5 276⫾13.3


LVEDD, mm 4.1⫾0.11 4.46⫾0.12* 3.7⫾0.11† 3.45⫾0.12
LVESD, mm 2.7⫾0.09 3.6⫾0.07* 1.9⫾0.06† 1.55⫾0.07
LVPWD, mm 0.7⫾0.012 0.8⫾0.012* 0.7⫾0.008 0.69⫾0.014
ES, % 46.3⫾1.2 29.4⫾1.3* 48.9⫾1.8 55.2⫾2.5
PAV, cm/s 144⫾5.5 117⫾3.2* 103⫾4.4† 101⫾5.8
CO, ml/min 51.7⫾1.5 36.46⫾1.9* 40.3⫾2.2† 32.2⫾2.4
LVM, mg 105⫾2.9 135⫾2.5* 80⫾1.5† 87⫾2.0
BW, gm 30⫾2.6 32⫾1.2 24⫾1.4 27⫾1.6
LVM/BW, mg/g 3.5⫾0.15 4.2⫾0.12* 3.8⫾0.11 3.7⫾0.17
Values are means ⫾ SE; n ⫽ 7 mice/group. HR, heart rate; LVEDD, left ventricle end-diastolic diameter; LVESD, left ventricle end-systolic diameter;
LVPWD, left ventricle posterior wall thickness; FS, fractional shortening; PAV, peak aortic velocity; CO, cardiac output; LVM, left ventricular mass; BW, body
weight. *P ⬍ 0.01 ANG II-treated group compared with respective vehicle-treated animals. †P ⬍ 0.01 poly(ADP-ribose)polymerase (PARP⫺/⫺) mice compared
with vehicle-treated PARP⫹/⫹ animals.

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org


H1548 HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II

Angiotensin infusion produces markedly reduced fibrosis in shown in Fig. 3B, cardiac NAD content reduced significantly
PARP⫺/⫺ mice. Impaired cardiac functions of a hypertrophied (30%) in angiotensin II-infused control (PARP⫹/⫹) mice but
heart and progression to cardiomyopathy are accompanied with not in mice lacking PARP gene. To further confirm these
disorganization of myocytes and increased interstitial fibrosis. results, we measured NAD content of primary cultures of
We, therefore, performed histological analysis to examine cardiac myocytes obtained from PARP⫹/⫹ and PARP⫺/⫺ mice.
cardiac remodeling response. Heart sections of control and Angiotensin II treatment significantly reduced the NAD con-
PARP⫺/⫺ mice were stained with ␣-actinin antibody and tent of PARP⫹/⫹ cardiomyocytes in a time-dependent manner.
Masson trichrome dye to visualize the organization of myofi- Maximum decline of NAD content was observed at 48 h after
bers and collagen content, respectively. As shown in Fig. 2, A angiotensin II treatment, when nearly 40% of cellular NAD
and B, in control hearts (PARP⫹/⫹), angiotensin II infusion content was lost. In contrast, in PARP⫺/⫺ cardiomyocytes, no
resulted in a disorganization of myofibers and increased car- significant decrease of NAD content was observed due to
diac fibrosis. Quantification of interstitial fibrosis showed a angiotensin II treatment (Fig. 3C). These results together dem-
marked increase in collagen deposition in hearts of angiotensin onstrated that in control (wild-type) cells, angiotensin II treat-
II-infused PARP⫹/⫹ mice. Contrary to this, in PARP⫺/⫺ mice, ment activates PARP, which in turn decreases cellular NAD
no significant change in cardiac fibrosis or alteration in myo- levels.
fibers organization was observed. These data indicated that the Evolution and progression of cardiac hypertrophy are ac-
cardiac remodeling response of angiotensin II is, in part, companied with increased protein synthesis and loss of myo-
contributed to by the presence of the intact PARP gene.
cyte survivability. We therefore determined the effect of an-
Angiotensin II infusion depletes cardiac NAD content of

Downloaded from ajpheart.physiology.org on March 16, 2009


giotensin II on protein synthesis by measuring incorporation of
PARP⫹/⫹ but not PARP⫺/⫺ mice. To demonstrate whether
[3H]leucine into total cell protein. PARP⫹/⫹ and PARP⫺/⫺
cardiac remodeling response of angiotensin II was indeed
contributed to by the activation of the PARP gene, we mea- myocytes were treated with a hypertrophy dose of angiotensin
sured poly-ADP-ribosylation of nuclear proteins. Cardiac nu- II (1 ␮M) and labeled with [3H]leucine for 72 h. Afterward,
clear extract of angiotensin II-infused PARP⫹/⫹ and PARP⫺/⫺ cells were harvested, total cell protein was precipitated, and the
mice was analyzed by Western blot analysis using anti-(ADP- incorporation of [3H]leucine was measured. As shown in Fig.
ribose) antibody. As shown in Fig. 3A, poly-ADP-ribosylation 4A, when compared with the vehicle-treated cells, [3H]leucine
of cardiac nuclear proteins was increased in angiotensin II- incorporation was significantly higher in PARP⫹/⫹ myocytes
infused PARP⫹/⫹ mice but not in PARP knockout mice, after angiotensin II treatment but not in cardiomyocytes defi-
compared with vehicle-treated controls, suggesting that PARP cient of the PARP gene. We next measured the impact of
is a target of angiotensin II-mediated signaling contributing to angiotensin II treatment on myocyte cell survivability by treat-
cardiac hypertrophy and failure. ing cells with higher doses of angiotensin II (10 and 20 ␮M).
PARP activation induces poly-ADP-ribosylation of proteins After 48 h of treatment, cells were stained with Hoechst and
by transferring multiple ADP-ribose moieties from NAD to the propidium iodide dyes, and cell death was determined by
target protein. This process consumes cellular NAD levels. FACS analysis. As shown in Fig. 4B, angiotensin II treatment
Therefore, a substantial drop in cellular NAD content is also induced a significant amount (30%) of cell death of PARP⫹/⫹
taken as a sign of PARP activation (18). To determine whether myocytes, as expected. However, no significant cell death of
cellular NAD levels were altered after stimulation of hearts PARP⫺/⫺ myocytes was observed after angiotensin II treat-
with angiotensin II, we measured total heart NAD content. As ment. These results indicated that angiotensin II-mediated

Fig. 2. Effect of ANG-II treatment on myo-


fibrillar organization and interstitial fibrosis
in PARP⫹/⫹ and PARP⫺/⫺ mice. A: repre-
sentative heart section (⫻200) immuno-
stained with ␣-actinin antibody and pro-
pidium iodide dye to visualize sarcomeres
and cell nuclei, respectively. B: interstitial
fibrosis was quantified from heart sections
(⫻100) stained with Masson’s trichrome
stain. Data presented as percent change of
collagen in ANG-II-treated wild-type
(PARP⫹/⫹) and PARP⫺/⫺ mice (n ⫽ 5, P ⬍
0.01).

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org


HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II H1549

Fig. 3. ANG II treatment increases PARP


activity in wild-type mice hearts, as mea-
sured by protein poly-ADP-ribosylation and
depletion of cellular NAD content. A: car-
diac nuclear extract of PARP⫹/⫹ and
PARP⫺/⫺ mice treated with vehicle (V) or
ANG II was analyzed by Western blot anal-
ysis using anti-poly(ADP-ribose) antibody.
Blots shown are representative of 4 experi-
ments. B: NAD content of the whole heart
cell lysate prepared from vehicle and ANG
II-treated PARP⫹/⫹ and PARP⫺/⫺ mice
(n ⫽ 4, *P ⬍ 0.05). C: primary cultures of
cardiac myocytes obtained from PARP⫹/⫹
and PARP⫺/⫺ mice were treated with 1 ␮M
of ANG II. At different time points of treat-
ment, cells were harvested, and NAD con-

Downloaded from ajpheart.physiology.org on March 16, 2009


tent was measured and presented as percent
change from untreated control (n ⫽ 3 at each
time point; *P ⬍ 0.05).

cardiac hypertrophy response and cell death were, in part, this, we treated cardiac myocytes with a hypertrophy dose of
attributed to PARP activation. different agonists, viz. isoproterenol, phenylephrine, and an-
We next asked whether PARP activation is a common target giotensin II. Forty-eight hours after treatment, cultures in
of different agonists producing cardiac hypertrophy. To test which cell hypertrophy was apparent under the microscope

Fig. 4. PARP⫺/⫺ cardiac myocytes are protected from ANG II-mediated hypertrophy and cell death. A: cardiomyocytes cultures prepared from PARP⫹/⫹ and
PARP⫺/⫺ mice were treated with ANG II (1 ␮M) and labeled with [3H]leucine. Seventy-two hours after treatment, cells were harvested and [3H]leucine
incorporation into total cell protein measured (n ⫽ 4 plates in each group; *P ⬍ 0.05). B: cardiac myocytes were treated with 10 or 20 ␮M of ANG II. Forty-eight
hours after treatment, cell death was measured by fluorescence-activated cell sorting analysis (n ⫽ 10 plates for each group; *P ⬍ 0.001). C: PARP⫹/⫹ myocytes
were treated with different hypertrophy agonists: isoproterenol (Iso), phenylephrine (Phe), and ANG II. For a positive control of PARP activation, cells were
treated with a free-radical generating mixture of H2O2 plus FeSO4 (0.1 mM each). Forty-eight hours after treatment, cells were harvested, and PARP activation
was measured using a PARP assay kit (see METHODS). Values are means ⫾ SE of 4 to 6 experiments.

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org


H1550 HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II

Fig. 5. NAD repletion and activation of Sir2␣ protect


PARP⫹/⫹ cardiomyocytes from ANG II-mediated cell
death. A: quantification of ANG II-mediated cell death
in different treatment groups, viz. cells overexpressing
NAD-biosynthetic enzyme nicotinamide phosphoribo-
syl-transferase (NAMPT), cells treated with 500 nM
resveratrol (Res), cells overexpressing Sir2␣, or res-
veratrol-treated (500 nM) cells in which Sir2␣ was
knocked out by small interfering RNA (siRNA). Open
bars, no ANG II treatment; gray bars, cells treated with
two different doses (10 and 20 ␮M) of ANG II for 48 h
(n ⫽ 5 experiments, *P ⬍ 0.05). Values are means ⫾

Downloaded from ajpheart.physiology.org on March 16, 2009


SE. B: NAD levels were determined in cells overex-
pressing NAMPT and/or treated with ANG II (20 ␮M).
Values are means ⫾ SE of 4 experiments. C: knock-
down of Sir2␣ expression by siRNA and overexpres-
sion by cell transfection was confirmed by Western blot
analysis using Sir2-specific antibody. Non-sp, nonspe-
cific reference band; Cont, control.

were harvested, and cell lysates were analyzed for the activity overexpression of NAD biosynthetic enzyme completely pro-
of poly-ADP-ribosylated proteins. Cells treated with a free tected cells against angiotensin II-mediated cell death. In these
radical-generating mixture of H2O2 and FeSO4 (0.1 mM each) experiments, we also measured cellular NAD content, and the
were used as positive control. As shown in Fig. 4C, all three results indicated that cellular NAD levels were in indeed
agonists stimulated PARP but with different degree of stimu- replenished by overexpression of the NAD biosynthetic en-
lation. Highest PARP stimulation was observed with angioten- zyme NAMPT (Fig. 5B). These data strongly indicated that
sin II, followed by phenylephrine and then isoproterenol treat- some of the detrimental effects (if not all) of angiotensin II in
ment. Angiotensin II-mediated stimulation of PARP activity PARP⫹/⫹ myocytes were contributed by PARP activation and
was comparable with that achieved from cells treated with free NAD depletion.
radical-generating mixture of H2O2 and FeSO4. These results Previous studies (3) have demonstrated that NAD is essen-
indicated that PARP activation could be a common down- tial for the activity of the longevity factor Sir2␣ deacetylase.
stream mechanism of different stimuli inducing hypertrophy, We therefore reasoned whether NAD depletion might have
and it plays a predominant role in angiotensin II-mediated resulted in the reduced activity of Sir2␣, and that, in fact, leads
cardiac hypertrophy. to cell death. To test this possibility, we treated PARP⫹/⫹
NAD repletion and Sir2␣ activation protects cardiac myo- myocytes with a Sir2␣ activator resveratrol before the addition
cytes from angiotensin II-mediated cell death. Having known of angiotensin II. As shown in Fig. 5, resveratrol pretreatment
that angiotensin II-mediated hypertrophy and cell death are markedly reduced angiotensin II-mediated cell death. Resvera-
associated with PARP activation and subsequent depletion of trol has been shown to have both antioxidant and Sir2␣
cellular NAD content, we next asked whether repletion of deacetylase-inducing activities (19). To determine which ac-
cellular NAD content could provide cell protection. To replen- tivity of resveratrol was effective in providing cell protection,
ish cellular NAD content, we transfected cells with a NAD we examined the effect of resveratrol in cells where Sir2␣
biosynthetic enzyme NAMPT. Overexpression of this enzyme levels were knocked down by siRNA. We found that reducing
increases cellular NAD content (25). To examine the effect of the Sir2␣ levels eliminated the protective effect of resveratrol,
this enzyme, PARP⫹/⫹ myocytes were transfected with the thus indicating that the beneficial effect of resveratrol was
NAMPT-expressing plasmid, and on the next day after trans- indeed attributed to Sir2␣ activation. To further confirm these
fection, cells were treated with angiotensin II (10 and 20 ␮M), results, we also examined the direct effect of Sir2␣ overex-
and cell death was determined 48 h later. As shown in Fig. 5, pression on survivability of PARP⫹/⫹ myocytes against angio-
AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org
HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II H1551
tensin II-induced cell death. As shown in Fig. 5, angiotensin stimulation was shown to trigger PARP activation in cultured
II-mediated cell death was drastically reduced in cells overex- endothelial cells and chronic infusion of angiotensin in mice
pressing Sir2␣ deacetylase. These results, together, demon- induced endothelial dysfunction, which was associated with
strated that PARP activation and subsequent depletion of NAD free radical production.
and the change in activity of Sir2␣ are the sequence of events One of the important features of cardiac hypertrophy and
contributing to the angiotensin II-mediated cardiac hypertro- remodeling is the accumulation of interstitial collagen. Previ-
phy and cell death. ously, overexpression of angiotensin receptor 1 in mice has
been shown to cause a significant increase of collagen depo-
DISCUSSION sition in the myocardium (22). Interestingly, the marked in-
crease in interstitial collagen content observed with angiotensin
PARP is known to be activated by cell oxidative stress (18). II infusion in wild-type mice was completely eliminated in
In this study we show that PARP is also involved in direct PARP knockout mice, indicating that PARP activation might
cardiac hypertrophy response of angiotensin II. A marked be the part of the process involved in the induction of collagen
attenuation of angiotensin II-induced cardiac hypertrophy was synthesis. This is consistent with previous reports (24, 32)
demonstrated in PARP knockout mice by measuring various where attenuation of interstitial fibrosis was observed in hearts
hypertrophy markers, including heart-to-tibia length ratio, of aortic-banded mice treated with PARP inhibitors or where
chamber dilation, myocyte cell size, interstitial fibrosis, and banding was carried out in mice deficient in the PARP gene.
expression of the fetal gene program. Results obtained from Our data obtained from cultured cardiac myocytes demon-
cultures of cardiac myocytes demonstrated that PARP⫺/⫺ strated that a hypertrophy dose of angiotensin II that induced

Downloaded from ajpheart.physiology.org on March 16, 2009


myocytes were protected from angiotensin II-dependent cell protein synthesis in control myocytes failed to produce similar
death. We also document a downstream mechanism of PARP changes in PARP⫺/⫺ myocytes. Although a direct role of
activation contributing to myocyte cell death and show that this PARP in protein synthesis is not yet known, there is strong
type of cell death could be prevented by activation of the evidence that PARP participates in regulation of general tran-
longevity factor Sir2␣ deactylase. These data provide the first scription and cardiac-specific muscle gene expression, includ-
in vivo evidence that PARP is a target of angiotensin II- ing expression of those genes that are known to be activated
mediated cell signaling contributing to cardiac hypertrophy, during hypertrophy (5, 28). Vyas et al. (28) have shown that
and the data support many previous reports where a beneficial PARP participates in transcription activation of ␤-MHC gene,
effect of PARP inhibitors was noticed in different experimental a hallmark of the fetal gene program that is activated during
models of heart failure (32). cardiac hypertrophy. Another important finding is that the dose
Angiotensin II is an established neurohumoral factor that of angiotensin II, which induced a significant amount of cell
induces cardiac hypertrophy. Multiple signal transduction death of PARP⫹/⫹ myocytes, failed to do so in myocytes
pathways have been demonstrated that converge the hypertro- devoid of the PARP gene, suggesting a role of PARP in
phy response of angiotensin II in cardiac myocytes. Recent angiotensin II-mediated myocytes cell demise. These results
reports have indicated that increased oxidative/nitrosative are consistent with previous reports (10, 18) where free radical-
stress is the major component of angiotensin II-mediated cell mediated necrosis of many cell types, such as neurons, thymo-
signaling contributing to hypertrophy of different cell types. In cytes, and pancreatic ␤-cells, was shown to be prevented by
vascular smooth muscle cells, endothelial cells as well as in PARP inhibitors or PARP deficiency.
cardiac myocytes angiotensin II have been shown to activate
membrane-bound NADPH oxidases, leading to increased pro-
duction of reactive oxygen and nitrogen species (2, 29). The
downstream mechanism of angiotensin II-mediated reactive
oxygen/nitrogen species production in cardiac myocytes is,
however, not known (until this study). In the present study,
mice were infused with a subpressor dose of angiotensin II.
This dose of angiotensin II produced nearly 25% cardiac
hypertrophy, which is associated with increased poly-ADP-
ribosylation of proteins. Consistent with this finding, a recent
report (2) has indicated that the same dose of angiotensin II,
which is used here, activates myocardial NADPH oxidase
activity, and the knocking down of NADPH-oxidase activity
attenuates hypertrophy response of angiotensin II in mice, thus
implicating that the dose used in this study was sufficient to
induce production of reactive oxygen and nitrogen species. In
addition, there is evidence showing that angiotensin II stimu-
Fig. 6. Schematic model showing involvement of PARP in ANG II-mediated
lation of cardiomyocytes causes DNA damage (14), which is cell death. Stimulation of cardiomyocytes with ANG II induces oxidative/
another profound stimulus of PARP activation. Our data pre- nitrosative stress. These signals activate PARP, leading to poly-ADP-ribosy-
sented here extend these previously reported intracellular cas- lation of proteins at expense of cellular NAD content. Depletion of cellular
cades of angiotensin II pathway and document that PARP is a NAD levels eventually leads to cell death. Our data show that this cascade of
events can be blocked (arrows with circle head) by NAD repletion, Sir2␣
downstream nuclear target of angiotensin II-mediated signal- activation, as well as by treatment of cells with resveratrol, which could protect
ing, leading to myocardial hypertrophy. These results are in cells by its antioxidative activity and/or by its ability to stimulate the activity
agreement with a recent report (27) where angiotensin II of Sir2␣ deacetylase.

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org


H1552 HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II

How does PARP activation induce hypertrophy and myocyte In conclusion, by using both in vitro and in vivo experi-
cell-death? PARP regulates cellular functions by both its en- ments, we have demonstrated that PARP is a nuclear target of
zymatic activity to induce protein poly-ADP-ribosylation as angiotensin II-mediated cell signaling leading to cardiac hy-
well as by its ability to bind to other proteins (4, 18, 20). Based pertrophy. A schematic model summarizing our results is given
on previous reports, the pathological effects of PARP activa- in Fig. 6. Our data support many previous studies where cell
tion are contributed by three main mechanisms. First, PARP oxidative/nitrosative stress was shown to be involved in the
activation catalyzes the transfer of multiple ADP-ribose moi- development of cardiac hypertrophy (reviewed in Ref. 21) and
eties from NAD to the target proteins, leading to depletion of in many other reports where failing hearts were found to be
cellular NAD content. This process eventually impairs the associated with increased activity of the PARP enzyme (1, 24,
energy metabolism and culminates in an energy deficit, leading 32). These results strengthen the notion that PARP inhibition
to cell dysfunction and death (4, 18). There is also evidence as monotherapy, or as a combination therapy that includes
that PARP activation causes poly-ADP-ribosylation and inac- activation of the Sir2␣, may represent a novel approach for the
tivation of the enzyme GAPDH, which could result in severe management of heart failure.
consequences to cell energy metabolism (9). Second, PARP
regulates gene transcription by chromatin remodeling as well ACKNOWLEDGMENTS
as by its ability to bind directly to other transcription factors (4, We thank Ayman Isbatan for technical assistance and the following inves-
5, 18). A plethora of evidence has indicated that PARP inter- tigators for their generous gift of different plasmids: Dr. V. Dawson (Depart-
acts with transcription factors, including YY-1, AP-2, AP-1, ment of Neurology, Johns Hopkins University School of Medicine, Baltimore,
MD) for PARP plasmid, Dr. W. Gu (Department of Pathology, College of
OCT-1, NF-␬B, p53, and TEF-1 and regulates the expression

Downloaded from ajpheart.physiology.org on March 16, 2009


Physician and Surgeons Columbia University, New York, NY) for Sir2
of several genes, including inducible nitric oxide synthase, plasmids, and Dr. Shin-Ichiro Imai (Department of Molecular Biology and
ICAM-1, and various cytokines and chemokines, and exerts a Pharmacology, Washington University School of Medicine, St. Louis, MO) for
profound inflammatory response (4, 18, 20). All these factors NAMPT plasmid.
have also been documented to participate in one or more steps
of the evolution and progression of cardiac hypertrophy. Third, GRANTS
PARP activation induces translocation of apoptosis-inducing This study was supported by National Heart, Lung, and Blood Institute
factor (AIF) from mitochondria to nucleus, leading to chro- Grants RO1-HL-68083 and RO1-HL-77788, American Heart Association
matinolysis and cell death without activation of caspases (34). Grant-in-aid N-150108, and the Department of Surgery research funds (to
M. P. Gupta).
Although each of these mechanisms is likely to contribute to
cardiac myocyte hypertrophy and cell death, depending on the REFERENCES
magnitude of PARP activation, our data presented here indi-
cate the presence of another new mechanism of PARP-medi- 1. Bartunek J, Vanderheyden M, Knaapen MW, Tack W, Kockx MM,
and Goethals M. Deoxyribonucleic acid damage/repair proteins are
ated cell death. We show that angiotensin II-mediated cell elevated in the failing human myocardium due to idiopathic dilated
death could be prevented by NAD repletion as well as by cardiomyopathy. J Am Coll Cardiol 40: 1097–1103, 2002.
treating cells with the Sir2␣ activator resveratrol. Knocking 2. Bendall JK, Cave AC, Heymes C, Gall N, and Ahah AM. Pivotal role
down Sir2␣ levels by siRNA eliminated the protective effect of of a gp91(phox)-containing NADPH oxidase in angiotensin II-induced
cardiac hypertrophy in mice. Circulation 105: 293–296, 2002.
resveratrol, thus indirectly indicating that PARP activation 3. Blander G and Guarente L. The Sir2 family of protein deacetylases.
threatens cell survival by reducing the activity of NAD-depen- Annu Rev Biochem 73: 417– 435, 2004.
dent class-III histone deacetylases (sirtuins). These findings are 4. Burkle A. Physiology and pathophysiology of poly(ADP-ribosyl)ation.
consistent with two earlier reports where a protective effect of Bioassays 23: 795– 806, 2001.
resveratrol treatment was observed against angiotensin II- 5. Butler AJ and Ordahl CP. Poly(ADP-ribose) polymerase binds with
transcription enhancer factor 1 to MCAT1 elements to regulate muscle-
mediated hypertrophy of cardiac and smooth muscle cells (6, specific transcription. Mol Cell Biol 19: 296 –306, 1999.
15). Prevention of angiotensin II-mediated cell death by Sir2 6. Cheng TH, Liu JC, Lin H, Shih NL, Chen YL, Huang MT, Chan P,
overexpression indicates that in these cells sufficient levels of Cheng CF, and Chen JJ. Inhibitory effect of resveratrol on angiotensin
cellular NAD must be available for the deacetylase reaction to II-induced cardiomyocyte hypertrophy. Naunyn Schmiedebergs Arch
Pharmacol 369: 239 –244, 2004.
go on. Consistent with this finding, we have recently shown 7. Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B,
that Sir2 overactivation prevents NAD loss, even in those cells Howitz KT, Gorospe M, Cabo Rde, and Sinclair DA. Calorie restriction
where PARP is ectopically overexpressed (23). Although the promotes mammalian cell survival by inducing the SIRT1 deacetylase.
precise mechanism of this effect is not known, one possible Science 305: 390 –392, 2004.
explanation could be that the Sir2 overexpression blocks the 7a.De Flora A, Zocchi E, Guida L, Franco L, and Bruzzone S. Autocrine
paracrine calcium signaling by the CD38/NAD⫹/cyclic ADP-ribose sys-
excessive enzymatic activity of PARP; as a result, NAD levels tem. Ann NY Acad Sci 1028: 176 –191, 2004.
are protected. Future studies are needed to figure out the 8. Devic E, Xiang Y, Gould D, and Kobilka B. ␤-Adrenergic receptor
possible interplay between these two molecules. If this possi- subtype-specific signaling in cardiac myocytes from ␤1 and ␤2 adreno-
bility turns out to be true, it will disclose that these two ceptor knockout mice. Mol Pharmacol 60: 577–583, 2001.
oxidative sensors reciprocally regulate the activity of each 9. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szabo C,
and Brownlee M. Inhibition of GAPDH activity by poly(ADP-ribose)
other to control the cell fate. In this context, it is worth noting polymerase activates three major pathways of hyperglycemic damage in
that there are reports suggesting that PARP and Sir2␣ exert an endothelial cells. J Clin Invest 112: 1049 –1057, 2003.
opposite effect on many apoptosis effectors, e.g., p53, Ku70, 10. Eliasson MJ, Sampei K, Mandir AS, Hurn PD, Traystman RJ, Bao J,
and NF-␬B. Whereas PARP activation alone or in conjunction Pieper A, Wang ZQ, Dawson TM, Snyder SH, and Dawson VL.
Poly(ADP-ribose) polymerase gene disruption renders mice resistant to
with other factors enhances their proapoptotic activity, Sir2␣ cerebral ischemia. Nat Med 3: 1089 –1095, 1997.
activation via deacetylation of the factor attenuates their apop- 12. Frey N, Katus HA, Olson EN, and Hill JA. Hypertrophy of the heart: a
totic potential (7, 13, 16, 26, 31, 33). new therapeutic target. Circulation 109: 1580 –1589, 2004.

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org


HYPERTROPHY RESPONSE OF PARP⫺/⫺ MICE TO ANGIOTENSIN II H1553
13. Galande S and Kohwi-Shigematsu T. Poly(ADP-ribose) polymerase and caspase-independent myocyte cell death during heart failure. Am J Physiol
Ku autoantigen form a complex and synergistically bind to matrix attach- Heart Circ Physiol 288: H486 –H496, 2005.
ment sequences. J Biol Chem 274: 20521–20528, 1999. 25. Revollo JR, Grimm AA, and Imai SI. The NAD biosynthetic pathway
14. Grishko V, Pastukh V, Solodushko V, Gillespie M, Azumo J, and mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activ-
Shaffer S. Apoptotic cascade initiated by angiotensin II in neonatal ity in mammalian cells. J Biol Chem 279: 50754 –50763, 2004.
cardiomyocytes: role of DNA damage. Am J Physiol Heart Circ Physiol 26. Smith JS. Human Sir2 and the silencing of p53 activity. Trends Cell Biol
285: H2364 –H2372, 2003. 12: 404 – 406, 2002.
15. Haider UG, Sorescu D, Griendling KK, Vollmar AM, and Dirsch VM. 27. Szabo C, Pacher P, Zsengeller Z, Vaslin A, Komjati K, Benko R, Chen
Resveratrol suppresses angiotensin II-induced Akt/protein kinase B and M, Mabley JG, and Kollai M. Angiotensin II-mediated endothelial
p70 S6 kinase phosphorylation and subsequent hypertrophy in rat aortic dysfunction: role of poly(ADP-ribose) polymerase activation. Mol Med
smooth muscle cells. Mol Pharmacol 62: 772–777, 2002. 10: 28 –35, 2004.
16. Hassa PO and Hottiger MO. The functional role of poly(ADP-ribose) 28. Vyas DR, McCarthy JJ, Tsika GL, and Tsika RW. Multiprotein
polymerase-1 as novel coactivator of NF-␬B in inflammatory disorders. complex formation at the beta myosin heavy chain distal muscle CAT
Cell Mol Life Sci 59: 1534 –1553, 2002. element correlates with slow muscle expression but not mechanical over-
17. Homburg S, Visochek L, Moran N, Dantzer F, Priel E, Asculai E, load responsiveness. J Biol Chem 276: 1173–1184, 2001.
Schwartz D, Rotter V, Dekel N, and Cohen-Armon M. A fast signal 29. Wang HD, Xu S, John DG, Du Y, Quinn MT, Cayatte AJ, and Cohen
induced activation of poly(ADP-ribose) polymerase: a novel down stream RA. Role of NADPH oxidase in the vascular hypertrophy and oxidative
target of phospholipase c. J Cell Biol 150: 293–307, 2000. stress response to angiotensin II in mice. Circ Res 88: 947–953, 2001.
18. Jagtap P and Szabo C. Poly(ADP-ribose) polymerase and the therapeutic 30. Wang Y. Signal transduction in cardiac hypertrophy dissecting compen-
effect of its inhibitors. Nat Rev Durg Discov 4: 421– 440, 2005. satory versus pathological pathways utilizing a transgenic approach. Curr
19. Kaeberlein M, McDonagh T, Heltweg B, Hixon J, Westman EA, Opin Pharmacol 1: 134 –140, 2001.
Caldwell SD, Napper A, Curtis R, DiStefano PS, Fields S, Bedalov A, 31. Wieler S, Gagne J, Vaziri H, Poirier GG, and Benchimol S. Poly(ADP)
and Kennedy BK. Substrate-specific activation of sirtuins by resveratrol. ribose polymerase-1 is a positive regulator of the p53-mediated G1 arrest

Downloaded from ajpheart.physiology.org on March 16, 2009


J Biol Chem 280: 17038 –17045, 2005. response after ionizing radiation. J Biol Chem 278: 18914 –18921, 2003.
20. Kraus WL and Lis JT. PARP goes transcription. Cell 113: 677– 683, 32. Xiao CY, Chen M, Zsengeller Z, Li H, Kiss L, Kollai M, and Szabo C.
2003. Poly(ADP-ribose) polymerase promotes cardiac remodeling, contractile
21. Pacher P, Schulz R, Liaudet L, and Szabo C. Nitrosative stress and failure, and translocation of apoptosis-inducing factor in a murine exper-
pharmacological modulation of heart failure. Trends Pharmacol Sci 26: imental model of aortic banding and heart failure. J Pharmacol Exp Ther
302–310, 2005. 312: 891– 898, 2005.
22. Paradis P, Dali-Youcef N, Paradis FW, Thibault G, and Nemer M. 33. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA,
Overexpression of angiotensin II type 1 receptor in cardiomyocytes and Mayo MW. Modulation of NF-␬B-dependent transcription and cell
induces cardiac hypertrophy and remodeling. Proc Natl Acad Sci USA 97: survival by the SIRT1 deacetylase. EMBO J 23: 2369 –2380, 2004.
931–936, 2000. 34. Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ,
23. Pillai JB, Isbatan A, Imai S, and Gupta MP. Poly(ADP-ribose) poly- Poirier GG, Dawson TM, and Dawson VL. Mediation of PARP-
merase-1-dependent cardiac myocyte cell death during heart failure is dependent cell death by apoptosis-inducing factor. Science 297: 259 –263,
mediated by NAD⫹ depletion and reduced Sir2alpha deacetylase activity. 2002.
J Biol Chem 280: 43121– 43130, 2005. 35. Zong WX, Ditsworth D, Bauer DE, Wang ZQ, and Thompson CB.
24. Pillai JB, Russell HM, Raman J, Jeevanandam V, and Gupta MP. Alkylating DNA damage stimulates a regulated form of necrotic cell
Increased expression of poly(ADP-ribose) polymerase-1 contributes to death. Genes Dev 18: 1272–1282, 2004.

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

You might also like