functional interaction between the transcription factor krüppel-like factor 5 and poly(adp-ribose)

16
1 FUNCTIONAL INTERACTION BETWEEN THE TRANSCRIPTION FACTOR KRÜPPEL-LIKE FACTOR 5 AND POLY(ADP-RIBOSE) POLYMERASE-1 IN CARDIOVASCULAR APOPTOSIS Toru Suzuki 1,2,5 , Toshiya Nishi 1,5 , Tomoko Nagino 1,5 , Kana Sasaki 1,5 , Kenichi Aizawa 1 , Nanae Kada 1 , Daigo Sawaki 1 , Yoshiko Munemasa 1 , Takayoshi Matsumura 1 , Shinsuke Muto 1,3 , Masataka Sata 1 , Kiyoshi Miyagawa 4 , Masami Horikoshi 3 and Ryozo Nagai 1 From the 1 Department of Cardiovascular Medicine, 2 Department of Clinical Bioinformatics, 4 Department of Radiation Biology, Graduate School of Medicine, and 3 Laboratory of Developmental Biology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, JAPAN 5 These authors contributed equally Running Title: Functional interaction between KLF5 and PARP-1 Address correspondence to: Toru Suzuki or Ryozo Nagai, Department of Cardiovascular Medicine, and Department of Clinical Bioinformatics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, JAPAN. Tel. 81-3-3815-5411 x33117; Fax. 81-3-5800-8824; E-mail. [email protected] or [email protected] Krüppel-like factor 5 (KLF5) is a transcription factor important in regulation of the cardiovascular response to external stress. KLF5 regulates pathological cell growth, and its acetylation is important for this effect. Its mechanisms of action, however, are still unclear. Analysis in KLF5-deficient mice showed that KLF5 confers apoptotic resistance in vascular lesions. Mechanistic analysis further showed that it specifically interacts with Poly(ADP-ribose) polymerase-1 (PARP-1), a nuclear enzyme important in DNA repair and apoptosis. KLF5 interacted with a proteolytic fragment of PARP-1, and acetylation of KLF5 under apoptotic conditions increased their affinity. Moreover, KLF5 wild-type but not a non-acetylatable point-mutant inhibited apoptosis as induced by the PARP-1 fragment. Collectively, we find that KLF5 regulates apoptosis, and to target PARP-1, and further for acetylation to regulate these effects. Our findings thus implicate functional interaction between the transcription factor KLF5 and PARP-1 in cardiovascular apoptosis. The cardiovasculature adapts dynamically to metabolic and/or mechanical stresses (i.e. blood vessel remodeling in response to oxidative and hypertensive stress). Although this response initially compensates for the pathological stimulus, chronic and excessive load ultimately leads to decompensatory maladaptation which is the underlying pathology of heart failure and atherosclerosis (1, 2). The cellular mechanisms underlying cardiovascular adaptation processes are characterized by cellular hyperplasia, hypertrophy and death. Previous studies have begun to clarify the molecular basis of the process centered on signaling pathways linking extracellular stimuli to intracellular processes characterized by the intracellular signaling cascade and downstream gene expression events, which include roles of transcription factors such as nuclear factor of activated T cells (NFAT) through the calcineurin pathway and histone deacetylases in cardiac hypertrophy (3-6). We have recently shown that the transcription factor, Krüppel-like factor 5 (KLF5), regulates the cardiovascular response to pathological stress (e.g. angiotensin II) by modulating atherosclerosis, angiogenesis and cardiac hypertrophy (7-10). http://www.jbc.org/cgi/doi/10.1074/jbc.M608098200 The latest version is at JBC Papers in Press. Published on February 5, 2007 as Manuscript M608098200 Copyright 2007 by The American Society for Biochemistry and Molecular Biology, Inc. by guest on December 31, 2018 http://www.jbc.org/ Downloaded from

Upload: others

Post on 23-Feb-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

1

FUNCTIONAL INTERACTION BETWEEN THE TRANSCRIPTION FACTOR KRÜPPEL-LIKE FACTOR 5 AND POLY(ADP-RIBOSE) POLYMERASE-1

IN CARDIOVASCULAR APOPTOSIS

Toru Suzuki1,2,5, Toshiya Nishi1,5, Tomoko Nagino1,5, Kana Sasaki1,5, Kenichi Aizawa1, Nanae Kada1, Daigo Sawaki1, Yoshiko Munemasa1, Takayoshi Matsumura1, Shinsuke Muto1,3,

Masataka Sata1, Kiyoshi Miyagawa4, Masami Horikoshi3 and Ryozo Nagai1

From the 1Department of Cardiovascular Medicine, 2Department of Clinical Bioinformatics, 4Department of Radiation Biology, Graduate School of Medicine, and

3Laboratory of Developmental Biology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, JAPAN

5These authors contributed equally Running Title: Functional interaction between KLF5 and PARP-1 Address correspondence to: Toru Suzuki or Ryozo Nagai, Department of Cardiovascular Medicine, and Department of Clinical Bioinformatics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, JAPAN. Tel. 81-3-3815-5411 x33117; Fax. 81-3-5800-8824; E-mail. [email protected] or [email protected] Krüppel-like factor 5 (KLF5) is a transcription factor important in regulation of the cardiovascular response to external stress. KLF5 regulates pathological cell growth, and its acetylation is important for this effect. Its mechanisms of action, however, are still unclear. Analysis in KLF5-deficient mice showed that KLF5 confers apoptotic resistance in vascular lesions. Mechanistic analysis further showed that it specifically interacts with Poly(ADP-ribose) polymerase-1 (PARP-1), a nuclear enzyme important in DNA repair and apoptosis. KLF5 interacted with a proteolytic fragment of PARP-1, and acetylation of KLF5 under apoptotic conditions increased their affinity. Moreover, KLF5 wild-type but not a non-acetylatable point-mutant inhibited apoptosis as induced by the PARP-1 fragment. Collectively, we find that KLF5 regulates apoptosis, and to target PARP-1, and further for acetylation to regulate these effects. Our findings thus implicate functional interaction between the transcription factor KLF5 and PARP-1 in cardiovascular apoptosis.

The cardiovasculature adapts dynamically to metabolic and/or mechanical stresses (i.e. blood vessel remodeling in response to oxidative and hypertensive stress). Although this response initially compensates for the pathological stimulus, chronic and excessive load ultimately leads to decompensatory maladaptation which is the underlying pathology of heart failure and atherosclerosis (1, 2).

The cellular mechanisms underlying cardiovascular adaptation processes are characterized by cellular hyperplasia, hypertrophy and death. Previous studies have begun to clarify the molecular basis of the process centered on signaling pathways linking extracellular stimuli to intracellular processes characterized by the intracellular signaling cascade and downstream gene expression events, which include roles of transcription factors such as nuclear factor of activated T cells (NFAT) through the calcineurin pathway and histone deacetylases in cardiac hypertrophy (3-6). We have recently shown that the transcription factor, Krüppel-like factor 5 (KLF5), regulates the cardiovascular response to pathological stress (e.g. angiotensin II) by modulating atherosclerosis, angiogenesis and cardiac hypertrophy (7-10).

http://www.jbc.org/cgi/doi/10.1074/jbc.M608098200The latest version is at JBC Papers in Press. Published on February 5, 2007 as Manuscript M608098200

Copyright 2007 by The American Society for Biochemistry and Molecular Biology, Inc.

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

2

While the transcriptional and signaling networks regulating the cardiac adaptation response have begun to be unraveled, further investigation is needed to better understand the pathogenic roles of the involved factors and pathways. Regulation of cell death/survival, in particular, remains poorly understood. Here, we show that KLF5 inhibits cell death/apoptosis and that it functionally interacts with Poly(ADP-ribose) polymerase-1 (PARP-1), a nuclear enzyme involved in the response to DNA damage (11).

MATERIALS and METHODS

Cell culture and apoptotic assays -

3T3 and HeLa cells were grown in DMEM supplemented with 10 % serum. Human umbilical vein-derived endothelial cells (HUVECs) were cultured in EBM-2 medium with EGM-2 supplement (Clonetics). Stable transformant cell lines derived from 3T3 and HeLa cells (12) were maintained in DMEM/10 % serum containing 50 µg/mL G418 (Sigma). For most cell death/survival experiments, cells were treated with 50 ng/mL recombinant TNF-α (Peprotech) and/or 4 µM actinomycin D (Sigma). Analysis in HUVECs was done following transfection of expression vectors. Caspase-3 was assayed with the Caspase-3 assay system (Promega). Cleaved DNA was assayed with the Cell death detection assay kit ELISA (Roche). TUNEL staining was done with the in situ apoptosis detection kit (Takara) after cells were fixed with 4 % paraformaldehyde. For the mouse femoral artery injury sections, the in situ death detection kit (Roche) was used. Nuclei were counterstained with propidium iodide (Sigma). Sections were mounted with ProLong Antifade Kit (Molecular Probes, Eugene, OR) and observed under a confocal microscope (FLUOVIEW FV300, Olympus, Tokyo). Mouse femoral artery injury model - Eight-week-old male KLF5 heterozygous knockout mice (9) and wild-type littermates were subjected to femoral artery injury and analyzed as described (13). RNA interference (RNAi) analysis - RNAi analysis of KLF5 was done as described

(14). Immunoprecipitation and Western blot analysis - Whole cell lysate or nuclear extract was immunoprecipitated with anti-FLAG M2 affinity gel (Sigma), prepared anti-KLF5 antibody or anti-PARP antibody (R&D systems) with protein G-Sepharose (GE Healthcare), subjected to SDS-PAGE analysis then immunoblotted as described (12, 14). For Western blot analysis, antibodies from the Apoptosis sampler kit (BD Biosciences) were used in addition to FLAG M2 monoclonal antibody (Sigma), anti-PARP-1 monoclonal antibody (BD Biosciences Pharmingen, R&D systems), anti-acetylated-lysine antibody (Santa Cruz) and anti-KLF5 antibody (KM1785)(9). Plasmid transfections were done using Lipofectamine 2000 (Invitrogen). Adenoviral transfections were done as described (14). Preparation of anti-KLF5 antibody - Anti-KLF5 antibody was prepared by immunizing rabbits with 100 µg of full-length purified recombinant 6HIS-KLF5 for 8 times at one week intervals after which serum was extracted. Antibody specificity was confirmed by Western blot (data not shown).

Preparation of recombinant epitope-tagged protein - Human KLF6 was PCR-amplified and subcloned into the pGEX vector (Amersham Pharmacia Biotech). Expression and purification of bacterial recombinant proteins for GST-KLF6, KLF5, KLF5-K369R, zinc fingers and 6HIS-24kDa PARP-1 were done essentially as described previously (12, 15, 16).

Protein-protein interaction assay and acetylation assay - Acetylation reactions and GST pull down assay were done as previously

described (12, 16). Statistical Analysis - All data were

analyzed by the non-paired t-test. p<0.05 was considered significant.

RESULTS Apoptotic resistance is a pathophysiological function of KLF5

The present study began with the initial observation of an attenuated response to vascular injury in KLF5 knockout mice

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

3

subjected to a mouse femoral artery injury model (9) (Figs. 1A, B). To understand the mechanisms underlying this effect on neointimal hyperplasia, we questioned whether cell death/apoptosis might play a pathological role given that cellular apoptosis is a major mechanism of the wire-injury model which was used (13). Analysis of apoptosis by TUNEL staining showed an increase in TUNEL-positive apoptotic cells in the neointima of knockout mice as compared to wild-type littermates (13.1% vs. 1.5%)(Figs. 1C, D). Insufficiency of KLF5 therefore resulted in decreased neointima formation most likely due to enhanced apoptotic cell death after vascular injury.

To confirm that KLF5 insufficiency is associated with increased apoptosis at the cellular level, we next used an RNA interference (RNAi) approach to knockdown KLF5 under apoptotic stimulation (tumor necrosis factor-α, TNF-α). TUNEL staining showed an increase in positive-staining cells when subjected to KLF5 siRNA as compared to control siRNA (secreted alkaline phosphatase, SEAP)(Figs. 1E, F). Thus, KLF5 insufficiency is associated with increased apoptosis. Cellular apoptotic resistance of KLF5

To characterize the apoptotic resistance mechanisms of KLF5, stable transformants (cloned) expressing KLF5 in human HeLa and murine 3T3 cells were used for further investigations. Both KLF5-expressing cells showed resistance to induced cell death by TNF-α as compared to mock cells (Fig. 2A). We then measured apoptotic caspase-3 activity which was reduced in both KLF5-expressing cells (29% for TNF-α stimulation and 44% for anisomycin treatment in HeLa cells, and 34% in 3T3 cells for TNF-α stimulation)(Fig. 2B). Further, TUNEL staining showed that KLF5-expressing cells were resistant to DNA-cleavage by apoptotic stimulation as shown by quantification of TUNEL-positive cells (15% by TNF-α and 40% by anisomycin) (Fig. 2C). Thus, cells expressing KLF5 were consistently resistant to apoptosis by criteria including morphology, caspase-3 activity and DNA cleavage (TUNEL).

Apoptotic resistance was confirmed in at least two independent clones for both HeLa and NIH3T3 cells as well as in a resistance-selected heterogenous non-cloned colony (data not shown). Mechanisms of apoptotic resistant activity of KLF5

Next, to investigate the molecular mechanisms of the apoptotic resistant effects of KLF5, expression levels of a panel of proteins related to the apoptotic signaling cascade were examined in the HeLa stable transformant with apoptotic stimulation by TNF-α. While there were no apparent effects on expression of most of these apoptosis-related proteins, the protein level of Poly(ADP-ribose) polymerase-1 (PARP-1) was markedly affected in KLF5-expressing cells as compared to control (Fig. 3A). PARP-1 is a 113 kDa nuclear enzyme involved in DNA repair that catalyzes the initiation, elongation, and branching of poly(ADP-ribose) onto its target protein (11). PARP-1 is cleaved by caspase into a 24 kDa amino-terminal DNA-binding domain and 89 kDa carboxyl-terminal catalytic domain under apoptotic conditions. This result was not unexpected given that caspase cleaves PARP-1 and as caspase-3 activity was reduced in KLF5-expressing cells (Fig. 2B). RNAi experiments confirmed involvement of PARP-1 in apoptosis of the tested HeLa cells (data not shown).

Nevertheless, we further pursued actions of KLF5 on PARP-1 under apoptotic conditions given the specific effects on PARP-1. We hypothesized that PARP-1 and KLF5 might functionally interact given that both KLF5 and PARP-1, notably its 24 kDa fragment, are zinc finger proteins/motifs which often physically and functionally interact (17). Immunoprecipitation experiments showed that KLF5 interacts with PARP-1, but strikingly for this interaction to be specific with the 24 kDa fragment under apoptotic conditions (Fig. 3B). Note that loading amounts of protein were normalized to show the difference in binding affinities. KLF5 did not interact with the 89 kDa carboxyl-terminal catalytic domain under these conditions (data not shown).

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

4

As these experiments were done using the stable transformant, further immunoprecipitation experiments were done to confirm interaction by endogenous proteins (Fig. 3C). Immunoprecipitation experiments using anti-KLF5 and PARP-1 antibodies confirmed that KLF5 and the PARP-1 24 kDa fragment interact in the cell. We thus sought to understand the functional implications and regulation of this interaction.

Interaction between KLF5 and PARP-1

To characterize the interaction between KLF5 and PARP-1, we next examined the specificity and site of interaction. For specificity, we compared binding of PARP-1 between KLF5 and KLF6, the latter being a similar Krüppel-like factor (18, 19) (Figs. 4A, B). GST pull-down assay under conditions in which KLF5 bound the 24 kDa fragment of PARP-1 (Fig. 4B, lane 4) showed lack of interaction with KLF6 (Fig. 4B, lane 5). Thus, interaction of KLF5 with PARP-1 was direct and specific.

We further determined the site of interaction between KLF5 and PARP-1. To confirm our initial expectations that the zinc finger motif is the protein-protein interaction interface (12, 17), we tested whether the zinc finger 24 kDa fragment of PARP-1 directly interacts with the zinc finger DNA-binding domain of KLF5 (Figs. 4C, D). GST pull-down assay showed that the zinc finger region of KLF5 directly and specifically bound the 24 kDa fragment of PARP-1 (Fig. 4D, lane 4). Given that the zinc finger regions of KLF5 and PARP-1 which mediate their interaction comprise their DNA-binding domains, we tested the requirement of DNA by competitively adding DNA to protein interaction assays which showed that DNA is not necessary for this interaction and for this interaction to thus be mediated by protein-protein interaction (data not shown).

As the zinc finger DNA-binding domain of KLF5 contains three zinc finger motifs, we next examined if there is specific binding of individual zinc fingers to PARP-1 (Fig. 4E). GST pull-down assay showed the first zinc finger but not the second nor third zinc

finger peptides to interact with PARP-1 (Fig. 4F, lanes 4-6). Acetylation is important for apoptotic resistant effects of KLF5

As interaction with PARP-1 was mediated through the first zinc finger of KLF5 (Fig. 4F) which contains a lysine residue whose acetylation we have previously shown to be important for the cell growth stimulatory effects of KLF5 (12), we next asked if acetylation is important for apoptotic resistant actions and interaction with PARP-1. First, we examined whether a non-acetylatable point-mutant of KLF5 (K369R, lysine to arginine substitution at residue 369) would lack resistance to apoptosis. Stable transformants in 3T3 cells of wild-type and that of the point-mutant K369R of KLF5 were subjected to apoptotic stimulus (TNF-α), and morphology was examined in addition to quantification of caspase-3 activity and DNA cleavage. As compared to wild-type KLF5 expressing cells, cells expressing the point-mutant KLF5-K369R were less viable in response to apoptotic stimulus (TNF-α)(Figs. 5A, B). Caspase-3 and DNA cleavage assay both showed that the point-mutant KLF5-K369R did not inhibit apoptosis under conditions in which KLF5 wild-type showed significant inhibition. Adenoviral transfer of the wild-type and point-mutant K369R into balloon-injured rat cartotid arteries confirmed that the wild-type but not the point-mutant K369R can inhibit pathophysiological vascular apoptosis (D. Sawaki and T. Suzuki et al., unpublished data). These findings suggest acetylation of KLF5 is important for its apoptotic resistant cellular effects.

The former experiments suggested that KLF5 is likely acetylated under apoptotic conditions. To test this, Western blot analysis using antibody against acetylated lysine was done which showed that KLF5 is markedly acetylated under apoptotic conditions (Fig. 5C, lane 3), although we did see some acetylation under basal conditions (lane 2). We next asked if acetylation might regulate interaction between KLF5 and the PARP-1 fragment. A protein-protein interaction assay using in vitro

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

5

acetylated KLF5 was done which showed that acetylation of KLF5 increased its binding affinity with the PARP-1 fragment (Fig. 5D, lane 4 vs. lane 3) although we do note that addition of p300 acetyltransferase region alone resulted in a marginal increase (data not shown). We further examined if acetylation augments interaction between KLF5 and the PARP-1 fragment in the cell. Immunoprecipitation experiments in endothelial cells transfected by adenovirus expressing similar amounts of wild-type or the point-mutant KLF5-K369R followed by immunoprecipitation of similar amounts of the 24 kDa PARP-1 fragment by anti-PARP-1 antibody showed that wild-type KLF5 but not the point-mutant KLF5-K369R to selectively interact with the PARP-1 24 kDa fragment (Fig. 5E, lane 2 vs. lane 3). Acetylation of KLF5 is thus induced under apoptotic conditions and is important for its apoptotic resistant activity as well as its interaction with PARP-1.

We further characterized the functional effects of this selective interaction. We reasoned that wild-type but not the point-mutant KLF5-K369R may inhibit apoptosis induced by the 24 kDa pro-apoptotic fragment of PARP-1 (16, 20). The 24 kDa pro-apoptotic fragment of PARP-1 and wild-type KLF5 or the point-mutant KLF5-K369R were transfected into endothelial cells and effects on apoptosis were determined by examining caspase-3 activity. Under conditions in which the 24 kDa pro-apoptotic fragment of PARP-1 stimulated apoptosis albeit marginally in our hands (Fig. 5F, lane 1 vs. lane 2), wild-type KLF5 inhibited caspase-3 activity in contrast to the point-mutant KLF5-K369R in which suppression of caspase-3 activity was not seen (Fig. 5F, lane 3 vs. lane 4). These experiments showed that wild-type KLF5 but not the point-mutant KLF5-K369R can inhibit apoptotic activity as stimulated by over-expression of the 24 kDa pro-apoptotic fragment of PARP-1.

DISCUSSION

Functional interaction between KLF5 and PARP-1

The cardiovascular transcription factor, Krüppel-like factor 5 (KLF5), inhibits cell death/apoptosis and functionally interacts with Poly(ADP-ribose) polymerase-1 (PARP-1), a highly abundant nuclear enzyme that functions as a sensor of DNA damage.

To our knowledge, KLF5 is the first protein to interact specifically and functionally with the 24 kDa PARP-1 fragment. This PARP-1 fragment also at least partially mediates physical interaction with the Werner syndrome protein, and also likely with DNA ligase III (21, 22), but neither the specificity nor the functional effect of the interaction had been addressed.

The PARP-1 fragment has been reported to harbor pro-apoptotic activity (16). We have found that KLF5 is able to inhibit the marginal pro-apoptotic effects of the PARP-1 fragment, that KLF5 interacts with this peptide and that acetylation of KLF5 stimulates this interaction. It is tempting to speculate that sequestration of the PARP-1 proteolytic fragment by KLF5 may be a novel target for regulation of PARP-1 actions.

However, we do note that the effects of PARP-1 on apoptosis remain controversial. Gene ablation studies in mice have shown that PARP-1 is not essential for apoptosis (23, 24). Additionally, cells exhibiting cleaved PARP-1 can divide normally (25) making its instructive role in apoptosis unclear. The fragment being produced after the apoptotic commitment step of caspase activation makes it further unlikely to be a critical determinant of apoptotic progression. Further investigation of the functional effect of the interaction with KLF5 will require a better understanding on the precise role of the PARP-1 fragment. Regulatory effects of acetylation

Another important finding of the present study is that the signaling modification, acetylation, was shown to play an important role in the effects of KLF5 on cell death and interaction with PARP-1. Acetylation is a nuclear-specific signaling modification which affects protein-protein as well as protein-DNA interactions by various nuclear factors (e.g. Armadillo and dTCF, Importin α and β)(26, 27). Although the biological role of this modification

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

6

is not well understood, we show that it affects multiple activities of KLF5.

A recent study showed that acetylation of Sp1, a close relative of KLF5 that we previously showed to be acetylated (28-30), can be similarly induced by an apoptosis-inducing anti-cancer agent (31), which together with our findings may suggest that acetylation plays a key role in regulation of cell death/survival pathways in this family of factors.

Further, as deacetylase (6) as well as acetylase and its activity (32) have been implicated in the cardiovascular remodeling response in the heart, this signaling pathway may have general implications for regulating the cardiovascular cell phenotype in response to pathological stress. KLF5 therefore through acting on apoptotic pathways may tip the balance between survival/repair and death/apoptosis under cardiovascular pathophysiological settings.

ACKNOWLEDGEMENTS

The authors thank Dr. M. Satoh for PARP-1 constructs. This study was supported by

grants from the Ministry of Education, Culture, Sports, Science and Technology, New Energy Development Organization, Ministry of Health, Labor and Welfare, and Japan Science and Technology Corporation.

REFERENCES 1. Olson, E. N. 2004. A decade of discoveries in cardiac biology. Nat. Med. 10: 467-474. 2. Towbin, J. A., and N. E. Bowles. 2002. The failing heart. Nature 415: 227–233. 3. Molkentin, J. D., J. R. Lu, C. L. Antos, B. Markham, J. Richardson, J. Robbins, S. R. Grant, and E. N. Olson. 1998. A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell 93: 215–228. 4. Molkentin, J. D., and I. G. Dorn II. 2001. Cytoplasmic signaling pathways that regulate cardiac hypertrophy. Annu. Rev. Physiol. 63:391–426. 5. Sussman, M. A., H. W. Lim, N. Gude, T. Taigen, E. N. Olson, J. Robbins, M. C. Colbert, A. Gualberto, D. F. Wieczorek, and J. D. Molkentin. 1998. Prevention of cardiac hypertrophy in mice by calcineurin inhibition. Science 281: 1690–1693. 6. Zhang, C. L., T. A. McKinsey, S. Chang, C. L. Antos, J. A. Hill, and E. N. Olson. 2002. Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell 110: 479–488. 7. Hoshino, Y., M. Kurabayashi, T. Kanda, A. Hasegawa, H. Sakamoto, E. Okamoto, K. Kowase, N. Watanabe, I. Manabe, T. Suzuki, A. Nakano, S. Takase, J. N. Wilcox, and R. Nagai. 2000. Regulated expression of the BTEB2 transcription factor in vascular smooth muscle cells; analysis of developmental and pathological expression profiles shows implications as a predictive factor for restenosis. Circulation 102: 2528-2534. 8. Shi, H., Z. Zhang, X. Wang, S. Liu, and C. T. Teng. 1999. Isolation and characterization of a gene encoding human Krüppel-like factor 5 (IKLF): binding to the CAAT/GT box of the mouse lactoferrin gene promoter. Nucleic Acids Res. 27: 4807-4815. 9. Shindo, T., I. Manabe, Y. Fukushima, K. Tobe, K. Aizawa, S. Miyamoto, K. Kawai-Kowase, N. Moriyama, Y. Imai, H. Kawakami, H. Nishimatsu, T. Ishikawa, T. Suzuki, H. Morita, K. Maemura,

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

7

M. Sata, Y. Hirata, M. Komukai, H. Kagechika, T. Kadowaki, M. Kurabayashi, and R. Nagai. 2002. Krüppel-like zinc-finger transcription factor KLF5/BTEB2 is a target for angiotensin II signaling and an essential regulator of cardiovascular remodeling. Nat. Med. 8: 856-863. 10. Watanabe, N., M. Kurabayashi, Y. Shimomura, K. Kawai-Kowase, Y. Hoshino, I. Manabe, M. Watanabe, M. Aikawa, M. Kuro-o, T. Suzuki, Y. Yazaki, and R. Nagai. 1999. BTEB2, a Krüppel-like transcription factor, regulates expression of the SMemb/nonmuscle myosin heavy chain B (SMemb/NMHC-B) gene. Circ. Res. 85:182-191. 11. D’Amours, D., S. Desnoyers, I. D’Silva, and G. G. Poirier. 1999. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem. J. 342: 249-268. 12. Miyamoto, S., T. Suzuki, S. Muto, K. Aizawa, A. Kimura, Y. Mizuno, T. Nagino, Y. Imai, N. Adachi, M. Horikoshi, and R. Nagai. 2003. Positive and negative regulation of the cardiovascular transcription factor KLF5 by p300 and the oncogenic regulator SET through interaction and acetylation on the DNA-binding domain. Mol. Cell. Biol. 23: 8528-8541. 13. Sata, M., Y. Maejima, F. Adachi, K. Fukino, A. Saiura, S. Sugiura, T. Aoyagi, Y. Imai, H. Kurihara, K. Kimura, M. Omata, M. Makuuchi, Y. Hirata, and R. Nagai. 2000. A mouse model of vascular injury that induces rapid onset of medial cell apoptosis followed by reproducible neointimal hyperplasia. J. Mol. Cell. Cardiol. 32: 2097-2104. 14. Aizawa, K., T. Suzuki, N. Kada, A. Ishihara, K. Kawai-Kowase, T. Matsumura, K. Sasaki, Y. Munemasa, I. Manabe, M. Kurabayashi, and R. Nagai. 2004. Regulation of Platelet-derived Growth Factor-A Chain by Kruppel-like Factor 5: new pathway of cooperative activation with nuclear factor-{kappa}B. J. Biol. Chem. 279: 70-76. J. Virol. 78: 8931-8934. 15. Suzuki, T., A. Kimura, R. Nagai, and M. Horikoshi. 2000. Regulation of interaction of the acetyltransferase region of p300 and the DNA-binding domain of Sp1 on and through DNA binding. Genes Cells 5: 29-41. 16. Yung, T. M. C., and M. S. Satoh. 2001. Functional competition between poly(ADP-ribose) polymerase and its 24-kDa apoptosis fragment in DNA repair and transcription. J. Biol. Chem. 276: 11279-11286. 17. Mackay, J. P., and M. Crossley. 1998. Zinc fingers are sticking together. Trends Biochem. Sci. 23: 1-4. 18. Narla, G., K. E. Heath, H. L. Reeves, D. Li, L. E. Giono, A. C. Kimmelman, M. J. Glucksman, J. Narla, F. J. Eng, A. M. Chan, A. C. Ferrari, J. A. Martignetti, and S. L. Friedman. 2001. KLF6, a candidate tumor suppressor gene mutated in prostate cancer. Science 294: 2563-2566. 19. Suzuki, T., T. Yamamoto, M. Kurabayashi, R. Nagai, Y. Yazaki, and M. Horikoshi. 1998. Isolation and initial characterization of GBF, a novel DNA-binding zinc finger protein that binds to the GC-rich binding sites of the HIV-1 promoter. J. Biochem. 124: 389-395. 20. Kim, J. W., J. Won, S. Sohn, and C.O. Joe. 2000. DNA-binding activity of the N-terminal cleavage product of poly(ADP-ribose) polymerase is required for UV mediated apoptosis. J. Cell Sci. 113: 955-961.

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

8

21. Schreiber, V., J. C. Amé, P. Dollé, I. Schultz, B. Rinaldi, V. Fraulob, J. Ménissier-de Murcia, and G. de Murcia. 2002. Poly(ADP-ribose) polymerase-2 (PARP-2) is required for efficient base excision DNA repair in association with PARP-1 and XRCC1. J. Biol. Chem. 277: 23028-23036. 22. von Kobbe, C., J. A. Harrigan, V. Schreiber, P. Stiegler, J. Piotrowski, L. Dawut, and V. A. Bohr. 2004. Poly(ADP-ribose) polymerase 1 regulates both the exonuclease and helicase activities of the Werner syndrome protein. Nucleic Acids Res. 32: 4003-4014. 23. De Murcia, J. M., C. Niedergang, C. Trucco, M. Ricoul, B. Dutrillaux, M. Mark, F. J. Oliver, M. Masson, A. Dierich, M. Le Meur, C. Walztinger, P. Chambon, and G. de Murcia. 1997. Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc. Natl. Acad. Sci. U. S. A. 94: 7303-7307. 24. Wang, Z. Q., L. Stingl, C. Morrison, M. Jantsch, M. Los, K. Schulze-Osthoff, and E. F. Wagner. 1997. PARP is important for genomic stability but dispensable in apoptosis. Genes Dev. 11: 2347-58. 25. Yang, L., Z. Cao, H. Yan, and W. C. Wood. 2003. Coexistence of high levels of apoptotic signaling and inhibitor of apoptosis proteins in human tumor cells: implication for cancer specific therapy. Cancer Res. 63: 6815-6824. 26. Bannister, A. J., E. A. Miska, D. Gorlich, and T. Kouzarides. 2000. Acetylation of importin-a nuclear import factors by CBP/p300. Curr. Biol. 10: 467-470. 27. Waltzer, L., and M. Bienz. 1998. Drosophila CBP represses the transcription factor TCF to antagonize Wingless signalling. Nature 395: 521-52. 28. Suzuki, T., S. Muto, S. Miyamoto, K. Aizawa, M. Horikoshi, and R. Nagai. 2003. Functional interaction of the DNA-binding transcription factor Sp1 through its DNA-binding domain with the histone chaperone TAF-I. J. Biol. Chem. 278: 28758-28764. 29. Suzuki, T., K. Aizawa, T. Matsumura, and R. Nagai. 2005. Vascular implications of the Kruppel-like family of transcription factors. Arterioscler. Thromb. Vasc. Biol. 25: 1135-1141. 30. Suzuki, T., T. Matsumura, and R. Nagai. 2005. Transcriptional regulation at the chromatin level in the cardiovasculature through protein-protein interactions and chemical modifications. Trends Cardiovasc Med. 15: 125-129. 31. Torigoe, T., H. Izumi, T. Wakasugi, I. Niina, T. Igarashi, T. Yoshida, I. Shibuya, K. Chijiiwa, K. Matsuo, H. Itoh, and K. Kohno. 2005. DNA topoisomerase II poison TAS-103 transactivates GC-box-dependent transcription via acetylation of Sp1. J. Biol. Chem. 280: 1179-1185. 32. Miyamoto, S., T. Kawamura, T. Morimoto, K. Ono, H. Wada, Y. Kawase, A. Matsumori, R. Nishio, T. Kita, and K. Hasegawa. 2006. Histone acetyltransferase activity of p300 is required for the promotion of left ventricular remodeling after myocardial infarction in adult mice in vivo. Circulation 113: 679-690.

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

9

FIGURE LEGENDS Fig. 1. Apoptotic resistance as a pathophysiological function of KLF5. A, Mouse femoral artery injury model in KLF5 wild-type (a, KLF5-wt) and heterozygous knockout (b, KLF5-KO+/-) mice. Hematoxylin and eosin staining. B, Intima to media (I/M) ratio (n=6). Error bars represent standard error. Scale bar, 50 µm; **, p<0.01. C, TUNEL staining of femoral artery injury samples. TUNEL is shown in green and propidium iodide (PI) in red. Arrow heads indicate merged TUNEL-positive nuclei (yellow). Arrows indicate the internal elastic lamina. Wild-type littermates (n=9) are shown in (a) and heterozygous knockout mice (n=8) are shown in (b); scale bar, 50 µm; NI, neointima; PC, phase contrast. D, Graphical representation of TUNEL-positive apoptotic cells. *, p<0.05. E, TUNEL-staining in apoptotic cells subjected to RNA interference. HeLa cells were used on the basis that they contain highest known endogenous levels of KLF5. G, Graphical representation of TUNEL-positive apoptotic cells. *, p<0.05. Fig 2. Apoptotic resistance activity of KLF5 in cells. A, Phase contrast photomicrographs of mock vector (HeLa, a-c; NIH-3T3, g-i) and KLF5 stable transformants (HeLa, d-f; NIH-3T3, j-l) treated with TNF-α. Time-course is as indicated. Scale bar, 100 µm. Quantification of cell survival shown on right. Results are presented as percent of control. **, p<0.01 vs. mock. B, Quantification of caspase-3 activity. Results are shown as percent of mock transfected cell line. **, p<0.01 vs. mock. C, TUNEL staining of cells. Photomicrographs of cells examined by phase contrast (PH) microscopy (a, b) and fluorescence microscopy (c, d) are shown. Apoptotic cells are identified by their highly condensed or fragmented nuclei (arrows in panel c). Quantification of DNA fragmentation shown on right. Results are shown as percent of mock transfected cell line. *, p<0.05. vs. mock; **, p<0.01 vs. mock. Fig 3. Mechanisms of KLF5 apoptotic resistance activity and effects on PARP-1. A, Immunoblots of apoptosis-related proteins in KLF5-expressing cells subjected to TNF-α apoptotic stimulus. TRADD, TNF-R-associated death domain; RIP, receptor interacting protein; FADD, Fas-associated death domain; Bad, Bcl-2-antagonist of cell death; Bax, Bcl-2-associated X protein; Bcl-2, B cell lymphoma/leukemia-2; DFF45, 45 kDa DNA fragmentation factor; PARP-1, poly(ADP-ribose) polymerase. B, Immunoprecipitation of apoptotic stimulus-induced 24 kDa pro-apoptotic fragment of PARP-1 by FLAG-tagged KLF5 in vivo. Lanes 1-4 are whole cell lysate input. Protein amounts were normalized to show the difference in binding affinities. Note that intact PARP-1 did not interact with KLF5 under these conditions. IP, immunoprecipitation; IB, immunoblot. C, Immunoprecipitation of endogenous KLF5 and PARP-1 proteins in endothelial cells. Immunoprecipitation by anti-KLF5 antibody followed by immunoblot with anti-PARP-1 antibody using pre-immune serum (PI) as control is shown on left, and the reverse experiment of immunoprecipitation by anti-PARP-1 antibody followed by immunoblot with anti-KLF5 (KM1785) antibody using IgG as control is shown on right. Cells were treated with anisomycin. Fig 4. Specific interaction between apoptotic resistant KLF5 and 24 kDa PARP-1 in vitro. A, Schematic representation of KLF5, KLF6 and PARP-1. B, GST pull-down assay of KLF5 and KLF6 with the 24 kDa pro-apoptotic fragment of PARP-1. C, Schematic representation of full-length KLF5 and DNA-binding domain (DBD) of KLF5. D, GST pull-down assay of KLF5 DNA-binding domain (DBD) and the 24 kDa pro-apoptotic fragment of PARP-1. E, Schematic representation of KLF5 zinc finger peptide motifs. F, GST pull-down assay of KLF5 zinc finger peptides and the 24 kDa pro-apoptotic fragment of PARP-1. Fig 5. Effect of acetylation on KLF5 apoptotic resistance and interaction with PARP-1. A, B, Effect of non-acetylatable point-mutant of KLF5 (K369R) on apoptotic resistance. K369R is a

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

10

lysine (K) to arginine (R) mutation at the 369th amino acid residue of KLF5. Morphological assessment by phase contrast microscopy (A) and quantification of caspase-3 activity and DNA cleavage (B). Results are shown as percent change of mock transfected cell line. *, p<0.05; **, p<0.01. C, Western blot analysis with anti-acetylated lysine-antibody under cellular apoptotic conditions. D, Effect of acetylation on interaction of KLF5 and PARP-1. Protein-protein interaction assayed using in vitro acetylated recombinant KLF5. E, Co-immunoprecipitation of the 24 kDa pro-apoptotic fragment of PARP-1 with wild-type KLF5 and point-mutant KLF5-K369R. F, Effects of KLF5 wild-type and K369R mutant on apoptosis as induced by the 24 kDa pro-apoptotic fragment of PARP-1 in endothelial cells. *, p<0.05 vs. mock.

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

Figure 1

% T

UNEL

pos

itive

cel

ls

0

5

10

20

15

KO+/-wtKLF5

1 2

DC

A

a bKLF5-wt KLF5-KO+/-

2.52.01.51.00.5

I/M ra

tio

KO+/-wtKLF5

1 20

**

B

*

0

2

4

6

% T

UNEL

pos

itive

cel

ls

KLF5ControlsiRNA

1 2

E

ba

KLF5 siRNAControl siRNA

*

F

KLF5-wtPC

TUNEL+PI

TUNEL

PI

Merged

PC PI

TUNELTUNEL+PIMerged

KLF5-KO+/-

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

Figure 2

Amock KLF5mock KLF5

control

TNF-α6hrs

TNF-α9hrs

NIH-3T3HeLa

a

b

c

d

e

f

g

h

i

j

k

l

Mock

Dead

cel

ls (%

of t

otal

)

KLF505

1015202530

1 2

Mock

Dead

cel

ls (%

of t

otal

)

KLF5

05

1015202530

1 2

HeLa

NIH-3T3

C

Mock

KLF5

PH TUNEL

a c

b dTNF-α Anisomycin

Mock KLF5 Mock KLF50

20406080

100120

1 2 3 4HeLa

Clea

ved

DNA

(% o

f moc

k)

B

Mock KLF5 Mock KLF5TNF-α AnisomycinHeLa Mock KLF5

TNF-αNIH-3T3

0

20

40

60

80

100

120

Casp

ase-

3 ac

tivity

(% o

f moc

k)

1 20

20

40

60

80

100120

1 2 3 4

Casp

ase-

3 ac

tivity

(% o

f moc

k)

**

**

****

***

**

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

Figure 3

C

A

1 2 1 2

TRADD

Moc

k

KLF5

DFF-45

RIP

Bcl-X

FADD

Bax

Caspase-2

Bad

Caspase-3

Caspase-7

Bcl-2 24 kDa PARP-1

Moc

k

KLF5

B

1 2 3 4 5 6 7 8

Mock KLF5TNF-α - +

Mock KLF5

IP

150 -100 -

75 -50 -37 -

25 -20 -

(kDa)

-PARP-1

- +-

+ - +++++-- -

- IP: FLAGIB: PARP-1

-24kDa PARP-1

-KLF5-24kDa PARP-1

1 2 3 4

PARP

-1Ig

G

KLF5

PI

IP:

IB: PARP-1 IB: KLF5

IP:

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

A

F

ResinInputGST-ZF1GST

ZF1,2

GST-ZF2

GST-ZF3

1 2 3 4 5 6

E

Activationdomain

DNA-bindingdomain

KLF5

KLF6

Zinc finger domain

Auto-modification

domainDNA-

bindingdomain

Catalyticdomain

24kDaPARP-1

PARP-1

C2H2 C2H2 C2H2ZF1

ZF2ZF3

CActivation

domainKLF5

KLF5-DBDZinc finger domain

DNA-bindingdomain

D

ResinInput

GST-KLF5-DBDGST

1 2 3 4

ZF3

ResinInputGST-KLF5

GST-KLF6GST

KLF5

1 2 3 4 5

B

Activationdomain

KLF5

DNA-bindingdomain

KLF6

24kDaPARP-1

24kDaPARP-1

Figure 4

24kDaPARP-1

-

-

-

-

-

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

Figure 5

AMock KLF5-wt KLF5-K369R

GST

GST-KLF5-DBDInput

p300-HAT+Ac-CoA ++ -

24kDa PARP-1

-

321 4

-

B

FLAG-KLF5Anisomycin

Ac-lysine

FLAG

IP FLAG

IB

3

+

++

2

-++

1

-+

-

-FLAG-KLF5

AcetylatedFLAG-KLF5-

a b c

C D

E F

0

40

80

120

1 2 3K369Rwt

KLF5Mock

Clea

ved

DNA

(% o

f moc

k)

**

060

80

140

1 2 3K369Rwt

KLF5Mock

Casp

ase-

3 ac

tivity

(% o

f moc

k)

***120

100

060

80

120

1 2 3K369Rwt

KLF5Mock

Casp

ase-

3 ac

tivity

(% o

f moc

k)

**

100

**

4

24kDa PARP-1

Ad-Empty wt

Ad-KLF5

KLF5

PARP-1

K369R

321

IB

PARP-1

24kDa PARP-1

Input

IP: KLF5

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from

Masataka Sata, Kiyoshi Miyagawa, Masami Horikoshi and Ryozo NagaiKada, Daigo Sawaki, Yoshiko Munemasa, Takayoshi Matsumura, Shinsuke Muto,

Toru Suzuki, Toshiya Nishi, Tomoko Nagino, Kana Sasaki, Kenichi Aizawa, Nanaepoly(ADP-ribose) polymerase-1 in cardiovascular apoptosis

Functional interaction between the transcription factor Kruppel-like factor 5 and

published online February 5, 2007J. Biol. Chem. 

  10.1074/jbc.M608098200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

by guest on Decem

ber 31, 2018http://w

ww

.jbc.org/D

ownloaded from