You are on page 1of 5

This article was downloaded by: [University of Otago]

On: 22 July 2015, At: 20:26


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG

Cell Cycle
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/kccy20

Hamlet
p18 Mediates Different p53-Dependent Responses
to DNA Damage Inducing Agents
Vanesa Lafarga, Ana Cuadrado & Angel R. Nebreda
Published online: 02 Oct 2007.

Hamlet
To cite this article: Vanesa Lafarga, Ana Cuadrado & Angel R. Nebreda (2007) p18 Mediates Different p53-Dependent
Responses to DNA Damage Inducing Agents, Cell Cycle, 6:19, 2319-2322, DOI: 10.4161/cc.6.19.4741

To link to this article: http://dx.doi.org/10.4161/cc.6.19.4741

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
[Cell Cycle 6:19, 2319-2322, 1 October 2007]; ©2007 Landes Bioscience

Extra View

p18Hamlet Mediates Different p53-Dependent Responses to DNA


Damage-Inducing Agents

Vanesa Lafarga Abstract


Ana Cuadrado* Cells organize appropriate responses to environmental cues by activating specific
signaling networks. Two proteins that play key roles in coordinating stress responses are
Angel R. Nebreda* the kinase p38α (MAPK14) and the transcription factor p53 (TP53). Depending on the
CNIO (Spanish National Cancer Center); Madrid, Spain nature and the extent of the stress-induced damage, cells may respond by arresting the
cell cycle or by undergoing cell death, and these responses are usually associated with
*Correspondence to: Angel R. Nebreda; CNIO (Spanish National Cancer Center);
Melchor Fernández Almagro 3; Madrid E-28029 Spain; Tel.: +34.91.2246900;
the phosphorylation of particular substrates by p38α as well as the activation of specific
Fax: +34.91.7328033; Email: anebreda@cnio.es/ Ana Cuadrado; CNIO (Spanish target genes by p53. We recently characterized a new p38α substrate, named p18Hamlet
National Cancer Center); Melchor Fernández Almagro 3; Madrid E-28029 Spain; (ZNHIT1), which mediates p53-dependent responses to different genotoxic stresses. Thus,
Tel.: +34.91.2246900; Fax: +34.91.7328033; Email: acuadrado@cnio.es cisplatin or UV light transiently stabilize of the p18Hamlet protein, which then enhances
Downloaded by [University of Otago] at 20:26 22 July 2015

Original manuscript submitted: 07/11/07 the ability of p53 to bind to and activate the promoters of pro-apoptotic genes such as
Manuscript accepted: 07/12/07 NOXA and PUMA leading to apoptosis induction. In a similar way, we report here that
Previously published online as a Cell Cycle E-publication:
p18Hamlet can also mediate the cell cycle arrest induced in response to γ-irradiation,
http://www.landesbioscience.com/journals/cc/article/4741 by participating in the p53-dependent upregulation of the cell cycle inhibitor p21Cip1
(CDKN1A).
Key words
p18Hamlet, p53, DNA damage, p21Cip1, cell
cycle, γ-radiation Introduction
Cells respond to extracellular stresses by activating specific intracellular signaling path‑
Acknowledgements
ways, such as the p38a mitogen‑activated protein kinase (MAPK) cascade, which in turn
V.L. acknowledges a pre-doctoral fellowship orchestrate different types of cellular responses, depending on the nature and strength of
from the Spanish Ministerio de Educacion the stimulus as well as the extent of the cellular damage. Typical cell fate responses induced
y Ciencia (MEC). A.R.N. is supported by by stresses that activate p38a are cell cycle arrest or apoptosis, which correlate with the
grants from MEC, Fundación La Caixa phosphorylation of a particular set of substrates in each case (reviewed in refs. 1 and 2).
and Fundación Científica de la Asociación The activation of p38a is one of the early events induced by the treatment of cancer
Española Contra el Cancer (AECC). cells with chemotherapeutic agents. In addition, p38 MAPKs can impinge on the
tumorigenic process at different levels (reviewed in refs. 3–5). Most available studies deal
with the function of p38a as a tumor suppressor, which may be accounted for by several
mechanisms such as the phosphorylation of p53 or the upregulation of the cell cycle
inhibitors p16Ink4a and p19Arf (see refs. 6–10). Recently, p38a has been shown to play
a key role in the negative regulation of malignant transformation induced by oncogenic
proteins that produce reactive oxygen species (ROS), such as H‑RasG12V. This inhibitory
effect is due to the ROS‑induced activation of p38a early in the process of transformation,
which induces apoptosis and prevents the accumulation of ROS and their carcinogenic
effects.11
Many papers support a role for p38a in the regulation of the tumor suppressor protein
p53. The majority of these studies propose a direct effect of either p38a or its target
MAPKAP kinase 2 (MK2) on the phosphorylation of several p53 serines (including
Ser15, Ser33, Ser46 and Ser392 for p38a, and Ser20 for MK2), which in turn positively
regulate p53 stability and activation.6,8 An additional mechanism that has been reported
is related to the ability of MK2 to phosphorylate the ubiquitin-ligase HDM2 on Ser157
and Ser166. Phosphorylation of these two residues appears to contribute to the activation
of HDM2 and therefore reduces p53 stability, modulating the extent and duration of the
stress‑induced p53 response.12
In an effort to identify new mediators of the p38a‑regulated stress responses, we
have recently characterized p18Hamlet as a substrate of p38a and p38b MAPKs that can
modulate p53 transcriptional activity.13 In fact, interfering with p18Hamlet expression
has an important inhibitory effect on the p53‑dependent apoptotic response to DNA

www.landesbioscience.com Cell Cycle 2319


p18Hamlet Mediates p21Cip1 Induction by p53

damage‑inducing agents of several human cancer cell lines as well


as primary mouse embryo fibroblasts (MEFs). However, p18Hamlet
does not seem to affect the stability of p53 or its phosphorylation on
Ser15 and Ser46, two of the main residues involved in the response
to DNA damage.
An extensive list of protein modifications has been reported
to contribute to p53 stabilization and activation, indicating an
enormous complexity in the mechanisms of p53 regulation.14,15
Thus, phosphorylation of several residues at the N‑terminus of p53
can regulate its interaction with HDM2 (and its mouse ortholog
MDM2), which targets p53 for ubiquitin‑mediated degradation.
In addition, the C‑terminus of p53 is rich in lysines, which are
subjected to modifications that can regulate the half‑life of the
p53 protein, such as acetylation, ubiquitination, sumoylation and
methylation.16,17
To further increase the complexity resulting from all these
post‑translational modifications, an additional mechanism of p53 Figure 1. p18Hamlet regulates p53 recruitment to the p21Cip1 promoter.
(A) U2OS cells with tetracycline inducible‑p18Hamlet were treated with tetra-
regulation has emerged in the last years as a collection of transcrip‑
cycline for 24 h or UV irradiated for 8 h and then subjected to ChIP analysis
tional coactivators that modulate p53 activity, apparently without using a p53 antibody (DO‑1 Santa Cruz) as described in ref. 13. The DNA
Downloaded by [University of Otago] at 20:26 22 July 2015

inducing any obvious modification in the p53 protein.18 This seems associated with the p53 immunoprecipitates was subjected to 32 cycles of
to be the case for the p38a‑regulated protein p18Hamlet, which PCR with primers corresponding to the CDKN1A and GAPDH promoters,
accumulates in response to several DNA damage agents and respectively. (B) U2OS cells were transfected with 100 nM of p18Hamlet or
control siRNAs (Dharmacon) and 72 h after transfection were g‑radiated
stimulates the recruitment of p53 to the promoter of certain target
(3 Gy) and then incubated for 6 h more before ChIP analysis. The DNA
genes, hence activating their transcription.13 associated with the p53 immunoprecipitates was amplified using CDKN1A
primers. Primers sequence is available upon request.
p18Hamlet as a Source for p53‑Dependent Response
Specificity
have been shown to differentially bind to particular promoters.
An interesting aspect of p18Hamlet is that it comprises several Thus, p53b (lacking the N‑terminal oligomerization domain) binds
features that may confer specificity to p53‑regulated stress responses. preferentially to the BAX and CDKN1A promoters, while the
First, in contrast to p38a and p53, which are ubiquitously expressed full‑length p53 binds better to the promoters of CDKN1A and
proteins, p18Hamlet mRNA levels are particularly high in tissues such MDM2 than to the BAX promoter.21 Determination of the p53
as spleen, kidney or brain, while almost undetectable in thymus, liver domain(s) involved in binding to coactivators should help to eluci‑
or testis.13 date whether association of these proteins with different p53 isoforms
Specificity can be also provided by the ability of the p18Hamlet accounts for promoter selectivity.
protein to accumulate in response to particular stress‑inducing
agents. After analyzing the behavior of p18Hamlet protein in response p18Hamlet is Involved in p21Cip1‑Mediated Cell Cycle
to various DNA damage‑inducing treatments, including both Arrest
physical agents and chemotherapeutic drugs, we concluded that
p18Hamlet protein levels increase only in response to a specific set of Our previous results suggested that p18Hamlet could specifically
treatments, such as cisplatin or UV radiation, but are refractory to enhance the p53‑dependent transcription of apoptosis‑inducing
others like etoposide and g‑radiation.13 At least in the case of UV, genes. This idea was based on the observation that, in response to
p18Hamlet accumulation depends on p38 MAPK activity, since this cisplatin or UV light treatments, the downregulation of p18Hamlet
effect is blocked by SB203580, an inhibitor of p38a and p38b. affected only the expression of pro‑apoptotic proteins, such as NOXA
However, it is possible that additional signaling pathways could also or PUMA, but not others such as p21Cip1 or HDM2. However, when
regulate p18Hamlet expression. The study of how p18Hamlet contrib‑ the effect of p18Hamlet on p53 recruitment to different promoters was
utes to different cellular responses that involve activation of the p38 analyzed using chromatin immunoprecipitation (ChIP), the results
MAPK pathway is an interesting field for future research. were somewhat different. Thus, although p18Hamlet overexpression
An additional level of specificity in p18Hamlet‑mediated cellular did not affect p21Cip1 protein levels,13 it substantially enhanced
responses may be related to its ability to stimulate transcription from p53 recruitment to the CDKN1A promoter (Fig. 1A). It should be
only certain p53 dependent promoters, such as those of NOXA or noted that, contrary to the behavior of other p53‑regulated proteins,
PUMA, but not from others such as HDM2. The same property has the p21Cip1 protein is not upregulated in many cell lines neither by
been previously reported for other p53 coactivators. For example, cisplatin nor UV light.13,22 Previous work has shown that although
ASPP proteins can stimulate the transcription of BAX and PIG3, but UV treatment induces a strong upregulation of p21Cip1 mRNA levels,
not of MDM2 or CDKN1A (see ref. 19) and the p300/CBP cofactor the p21Cip1 protein does not accumulate in UV‑treated cells and this
JMY can efficiently upregulate BAX but not CDKN1A (ref. 20). The degradation of p21Cip1 is essential for optimal DNA repair.22,23
mechanisms that determine promoter specificity of the p53 coactiva‑ It is known that p21Cip1 has a key role in p53‑induced cell cycle
tors have not been elucidated yet. However, different p53 isoforms arrest and human osteosarcoma U2OS cells are broadly used for the

2320 Cell Cycle 2007; Vol. 6 Issue 19


p18Hamlet Mediates p21Cip1 Induction by p53

Figure 3. Downregulation of p18Hamlet impairs the p21Cip1‑dependent G1


cell cycle arrest induced by g‑radiation. U2OS cells were transfected with
p18Hamlet or control siRNAs and 48 h after transfection were treated with
3 Gy of g‑radiation and then analyzed by flow cytometry at the indicated
times. The arrowhead indicates the establishment of the G1/S checkpoint that
takes place in control cells.
Downloaded by [University of Otago] at 20:26 22 July 2015

upregulation was significantly reduced (about 50%) in p18Hamlet‑


depleted cells (Fig. 2A and B). Similarly, g‑radiation‑induced p21Cip1
protein levels were affected to the same extent by the downregula‑
tion of p18Hamlet (Fig. 2C). To determine the biological relevance of
p18Hamlet in the establishment of the p21Cip1‑dependent G1 arrest,
we analyzed the behavior of both control and p18Hamlet‑depleted
cells in response to 3 Gy of g‑radiation. As shown in Figure 3, the G1
checkpoint was already established in control cells 12 h after g‑radi‑
ation, as indicated by the reduction in the cell population entering
into S phase (arrowhead). As a consequence, 16 h after the treatment,
Figure 2. p18Hamlet is required for p21Cip1 induction in response to g‑radia- there was still a significant percentage of cells that remained arrested
tion. (A) U2OS cells were transfected with p18Hamlet or control siRNAs and in G1. In contrast, downregulation of p18Hamlet impaired G1 arrest
48 h after transfection were treated with 3 Gy of g‑radiation. Total RNA
was isolated at the indicated times and the levels of p18Hamlet and p21Cip1
and cells continued to enter into S phase, accumulating mostly in
mRNAs were analyzed by Northern Blot using specific probes. Methylene G2/M.
blue staining of 28S rRNA was used to confirm equal RNA loading. The role of p18Hamlet in the control of the p53/p21Cip1‑mediated
(B) Quantification of p21Cip1 mRNA levels normalized to GAPDH. (C) U2OS cell‑cycle arrest that we describe here, together with the reported
cells were transfected with p18Hamlet or control siRNAs and 48 h after function of p18Hamlet as a mediator of p53‑dependent apoptosis,13
transfection were treated with 3 Gy or 6 Gy of g‑radiation. 24 h after the
treatment, p21Cip1 and p18Hamlet protein levels were analyzed by Western
strongly support the importance of p18Hamlet in the regulation of
blot. Tubulin was used as a loading control. this tumor suppressor protein. A possible model that summarizes
p18Hamlet function in the context of p53‑mediated responses is
shown in Figure 4. In response to DNA damage‑inducing agents,
study of the G1 checkpoint. In response to many DNA damage‑ such as cisplatin or UV light that induce strong and sustained p38a
inducing agents, including g‑radiation, these cells can arrest in G1 and activation, p18Hamlet becomes phosphorylated by p38a on several
G2 phases.24 Therefore, we used U2OS cells treated with g‑radiation threonine residues. These phosphorylations increase the half‑life
to clarify the contribution of p18Hamlet to the regulation of p21Cip1 of the p18Hamlet protein, which then accumulates and binds to
expression. Consistent with the above‑mentioned observation that p53, favoring the recruitment of this transcription factor to certain
p18Hamlet overexpression enhances the recruitment of p53 to the p53‑dependent promoters such as NOXA or PUMA that are involved
CDKN1A promoter, we also observed that the downregulation of in the apoptosis response. In contrast, g‑radiation establishes a
p18Hamlet significantly reduced p53 recruitment to this promoter in different scenario, in which the activation of p38a is rather weak
response to g‑radiation (Fig. 1B). This result suggested that p18Hamlet and transient (data not shown and ref. 27). Intriguingly, under
could potentially regulate the CDKN1A promoter in a similar way as these conditions, p38a activation does not appear to correlate with
the NOXA promoter in response to other types of DNA damage. p18Hamlet accumulation, although nevertheless p18Hamlet is still
Next, we analyzed the contribution of p18Hamlet to the G1 phase required for the recruitment of p53 to the CDKN1A promoter. This
cell cycle arrest induced by g‑radiation in U2OS cells, which is results in enhanced expression of the p21Cip1 protein, which in turn
known to depend on p21Cip1 accumulation.25,26 As expected, g‑radi‑ is responsible for the subsequent cell cycle arrest. The possible contri‑
ation rapidly induced the upregulation of p21Cip1 mRNA levels in bution of p38a to the p18Hamlet‑mediated upregulation of p21Cip1 in
U2OS cells transfected with control siRNA, whereas p21Cip1 mRNA response to g‑radiation remains to be elucidated.

www.landesbioscience.com Cell Cycle 2321


p18Hamlet Mediates p21Cip1 Induction by p53

References
1. Nebreda AR, Porras A. p38 MAP kinases: Beyond the stress response. Trends Biochem Sci
2000; 25:257‑60.
2. Ono K, Han J. The p38 signal transduction pathway: Activation and function. Cell Signal
2000; 12:1‑13.
3. Bulavin DV, Fornace Jr AJ. p38 MAP kinase’s emerging role as a tumor suppressor. Adv
Cancer Res 2004; 92:95‑118.
4. Bradham C, McClay DR. p38 MAPK in development and cancer. Cell Cycle 2006;
5:824‑8.
5. Dolado I, Nebreda AR. Regulation of tumorigenesis by p38a MAP kinase. Topics Current
Gen 2007; In press.
6. Bulavin DV, Saito S, Hollander MC, Sakaguchi K, Anderson CW, Appella E, Fornace Jr AJ.
Phosphorylation of human p53 by p38 kinase coordinates N‑terminal phosphorylation and
apoptosis in response to UV radiation. EMBO J 1999; 18:6845‑54.
7. Sanchez‑Prieto R, Rojas JM, Taya Y, Gutkind JS. A role for the p38 mitogen‑acitvated
protein kinase pathway in the transcriptional activation of p53 on genotoxic stress by che‑
motherapeutic agents. Cancer Res 2000; 60:2464‑72.
8. She QB, Chen N, Dong Z. ERKs and p38 kinase phosphorylate p53 protein at serine 15 in
response to UV radiation. J Biol Chem 2000; 275:20444‑9.
9. Bulavin DV, Phillips C, Nannenga B, Timofeev O, Donehower LA, Anderson CW, Appella
E, Fornace Jr AJ. Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis
through p38 MAPK‑mediated activation of the p16(Ink4a)‑p19(Arf ) pathway. Nat Genet
2004; 36:343‑50.
10. Timofeev O, Lee TY, Bulavin DV. A subtle change in p38 MAPK activity is sufficient to
suppress in vivo tumorigenesis. Cell Cycle 2005; 4:118‑20.
11. Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR. p38a MAP kinase as a
sensor of reactive oxygen species in tumorigenesis. Cancer Cell 2007; 11:191‑205.
Downloaded by [University of Otago] at 20:26 22 July 2015

12. Weber HO, Ludwig RL, Morrison D, Kotlyarov A, Gaestel M, Vousden KH. HDM2
phosphorylation by MAPKAP kinase 2. Oncogene 2005; 24:1965‑72.
13. Cuadrado A, Lafarga V, Cheung PC, Dolado I, Llanos S, Cohen P, Nebreda AR. A new p38
MAP kinase‑regulated transcriptional coactivator that stimulates p53‑dependent apoptosis.
Figure 4. A model for p18Hamlet function in p53‑dependent DNA damage EMBO J 2007; 26:2115‑26.
responses. Doses of cisplatin and UV that induce apoptosis in U2OS cells 14. Bode AM, Dong Z. Post‑translational modification of p53 in tumorigenesis. Nat Rev
activate the p38a MAPK pathway, which in turn phosphorylates p18Hamlet in Cancer 2004; 4:793‑805.
at least four threonines (Thr6, Thr64, Thr71, Thr103). As a consequence of 15. Appella E, Anderson CW. Post‑translational modifications and activation of p53 by geno‑
toxic stresses. Eur J Biochem 2001; 268:2764‑72.
these phosphorylations, p18Hamlet accumulates and stimulates the recruitment 16. Brooks CL, Gu W. Ubiquitination, phosphorylation and acetylation: The molecular basis
of p53 to the promoters of pro‑apoptotic genes, such as PUMA and NOXA, for p53 regulation. Curr Opin Cell Biol 2003; 15:164‑71.
with the subsequent induction of apoptosis. Interestingly, although p18Hamlet 17. Prives C, Manley JL. Why is p53 acetylated? Cell 2001; 107:815‑8.
does not appear to accumulate in response to doses of g‑radiation that induce 18. Coutts AS, La Thangue NB. The p53 response: Emerging levels of cofactor complexity.
G1 cell cycle arrest, this protein is still necessary for p53 recruitment to the Biochem Biophys Res Commun 2005; 331:778‑85.
19. Samuels‑Lev Y, O’Connor DJ, Bergamaschi D, Trigiante G, Hsieh JK, Zhong S, Campargue
CDKN1A promoter and for the corresponding activation of the p21Cip1‑
I, Naumovski L, Crook T, Lu X. ASPP proteins specifically stimulate the apoptotic function
dependent G1 checkpoint. The mechanisms underlying the role of p18Hamlet of p53. Mol Cell 2001; 8:781‑94.
in p21Cip1 upregulation as well as the possible contribution of p38a (or other 20. Shikama N, Lee CW, France S, Delavaine L, Lyon J, Krstic‑Demonacos M, La Thangue
protein kinases) to this process are still unknown. NB. A novel cofactor for p300 that regulates the p53 response. Mol Cell 1999; 4:365‑76.
21. Bourdon JC, Fernandes K, Murray‑Zmijewski F, Liu G, Diot A, Xirodimas DP, Saville
MK, Lane DP. p53 isoforms can regulate p53 transcriptional activity. Genes Dev 2005;
19:2122‑37.

A Role for p18Hamlet in Chromatin Remodeling 22. Bendjennat M, Boulaire J, Jascur T, Brickner H, Barbier V, Sarasin A, Fotedar A, Fotedar R.
UV irradiation triggers ubiquitin‑dependent degradation of p21(WAF1) to promote DNA
repair. Cell 2003; 114:599‑610.
Our results strongly support that p18Hamlet can regulate the 23. Fotedar R, Bendjennat M, Fotedar A. Role of p21WAF1 in the cellular response to UV. Cell
activity of p53, but the molecular mechanisms that underlie p18Hamlet Cycle 2004; 3:134‑7.
24. Berns K, Hijmans EM, Mullenders J, Brummelkamp TR, Velds A, Heimerikx M,
function remain to be elucidated. Interestingly, the role of p18Hamlet Kerkhoven RM, Madiredjo M, Nijkamp W, Weigelt B, Agami R, Ge W, Cavet G, Linsley
as a transcriptional coactivator is further supported by recent work, PS, Beijersbergen RL, Bernards R. A large‑scale RNAi screen in human cells identifies new
components of the p53 pathway. Nature 2004; 428:431‑7.
which has identified this protein as a subunit of the human SRCAP 25. Waldman T, Kinzler KW, Vogelstein B. p21 is necessary for the p53‑mediated G1 arrest in
(SNF2‑related CBP‑activating protein) complex.28 This complex is human cancer cells. Cancer Res 1995; 55:5187‑90.
26. Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW,
responsible for the exchange of histone H2A.Z in the promoters of Vogelstein B. Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science
certain genes in many different organisms, including plants, yeast 1998; 282:1497‑501.
27. Raman M, Earnest S, Zhang K, Zhao Y, Cobb MH. TAO kinases mediate activation of p38
and mammals.29‑31 At least in plants and yeast, p18Hamlet homo‑ in response to DNA damage. EMBO J 2007; 26:2005‑14.
logues have been shown to be essential for the function of the SRCAP 28. Cai Y, Jin J, Florens L, Swanson SK, Kusch T, Li B, Workman JL, Washburn MP, Conaway
complex.31,32 Interestingly, recent efforts to generate high‑resolution RC, Conaway JW. The mammalian YL1 protein is a shared subunit of the TRRAP/TIP60
histone acetyltransferase and SRCAP complexes. J Biol Chem 2005; 280:13665‑70.
maps for the human genome‑wide distribution of different histone 29. Mizuguchi G, Shen X, Landry J, Wu WH, Sen S, Wu C. ATP‑driven exchange of his‑
modifications, have determined that the histone H2A.Z variant tone H2AZ variant catalyzed by SWR1 chromatin remodeling complex. Science 2004;
303:343‑8.
is enriched in the promoter of active human genes.32,33 These 30. Deal RB, Topp CN, McKinney EC, Meagher RB. Repression of flowering in Arabidopsis
results suggest the possibility that p18Hamlet could regulate gene requires activation of FLOWERING LOCUS C expression by the histone variant H2A.Z.
Plant Cell 2007; 19:74‑83.
transcription through the remodeling of chromatin histones in 31. March‑Diaz R, Garcia‑Dominguez M, Florencio FJ, Reyes JC. SEF, a new protein required
certain promoters and in response to specific environmental cues. for flowering repression in Arabidopsis, interacts with PIE1 and ARP6. Plant Physiol 2007;
143:893‑901.
The potential implication of SRCAP in the p18Hamlet‑mediated 32. Wu WH, Alami S, Luk E, Wu CH, Sen S, Mizuguchi G, Wei D, Wu C. Swc2 is a widely
regulation of p53 and perhaps other transcription factors should be conserved H2AZ‑binding module essential for ATP‑dependent histone exchange. Nat
Struct Mol Biol 2005; 12:1064‑71.
an exciting issue for future research.
33. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao
K. High‑resolution profiling of histone methylations in the human genome. Cell 2007;
129:823‑37.

2322 Cell Cycle 2007; Vol. 6 Issue 19

You might also like