ArticlePDF Available

The ATM-related domain of TRRAP is required for histone acetyltransferase recruitment and Myc-dependent oncogenesis

Authors:

Abstract and Figures

The ATM-related TRRAP protein is a component of several different histone acetyltransferase (HAT) complexes but lacks the kinase activity characteristic of other ATM family members. We identified a novel function for this evolutionarily conserved domain in its requirement for the assembly of a functional HAT complex. Ectopic expression of TRRAP protein with a mutation in the ATM-related domain inhibits Myc-mediated oncogenic transformation. The Myc-binding region of TRRAP maps to a separable domain, and ectopic expression of this domain inhibits cell growth. These findings demonstrate that the ATM-related domain of TRRAP forms a structural core for the assembly and recruitment of HAT complexes by transcriptional activators.
Content may be subject to copyright.
RESEARCH COMMUNICATION
The ATM-related domain
of TRRAP is required for histone
acetyltransferase recruitment
and Myc-dependent oncogenesis
Jeonghyeon Park,
1
Sudeesha Kunjibettu,
2
Steven B. McMahon,
2
and Michael D. Cole
1,3
1
Department of Molecular Biology, Lewis Thomas Laboratory,
Princeton University, Princeton, New Jersey 08544-1014,
USA;
2
The Wistar Institute, Philadelphia,
Pennsylvania 19104-4268, USA
The ATM-related TRRAP protein is a component of sev-
eral different histone acetyltransferase (HAT) complexes
but lacks the kinase activity characteristic of other ATM
family members. We identified a novel function for this
evolutionarily conserved domain in its requirement for
the assembly of a functional HAT complex. Ectopic ex-
pression of TRRAP protein with a mutation in the ATM-
related domain inhibits Myc-mediated oncogenic trans-
formation. The Myc-binding region of TRRAP maps to a
separable domain, and ectopic expression of this domain
inhibits cell growth. These findings demonstrate that
the ATM-related domain of TRRAP forms a structural
core for the assembly and recruitment of HAT com-
plexes by transcriptional activators.
Received March 30, 2001; revised version accepted May 14,
2001.
Members of the ATM family of proteins function in hu-
mans, yeast, and other organisms as critical sensors of
genomic integrity or growth conditions. In humans, the
ATM family includes ATM, ATR, FRAP, and DNA-
PKcs, and homologs of these proteins are found in yeast,
flies, and other organisms (e.g., Mec1p, Tel1p, RAD3p,
TOR1p, and Mei-41; Keith and Schreiber 1995). The de-
fining features of the proteins in the ATM family are
their general large size (2500–4000 amino acids) and a
300-amino-acid motif in their C terminus that resembles
the catalytic domain of PI3-kinases. However, most of
these proteins have been shown to phosphorylate other
proteins rather than lipids. More extended alignment of
the ATM family members identifies two other conserved
domains in addition to the PI3-kinase-like domain (Bo-
sotti et al. 2000). The PI3-kinase-like domain is flanked
by the FATC domain of 35 amino acids at the extreme C
terminus and the FAT domain, which comprises 500
amino acids amino terminal of the catalytic domain. All
existing evidence suggests that the integrity of the ki-
nase domain is essential for the function of ATM family
proteins, but the functions of the FATC and FAT do-
mains remain unknown. However, our recent identifica-
tion of the novel ATM-related TRRAP proteins has
raised new questions about the function of these con-
served domains. The TRRAP protein family was identi-
fied as transcriptional cofactors that mediate the recruit-
ment of large multiprotein histone acetyltransferase
(HAT) complexes to sequence-specific activators (Grant
et al. 1998; McMahon et al. 1998; Saleh et al. 1998; Vas-
silev et al. 1998). We initially identified TRRAP or-
thologs in humans, Saccharomyces cerevisiae,Schizo-
saccharomyces pombe, and Caenorhabditis elegans, and
more recently in Arabidopsis thaliana and Drosophila
melanogaster. The TRRAP proteins are true members of
the ATM family because they possess the FAT, kinase-
related, and FATC domains. However, the kinase-related
domain is conserved in sequence but not in function as
the specific residues mediating phosphate transfer are
absent (McMahon et al. 1998). This lack of catalytic ac-
tivity has been supported by biochemical studies on both
the human and yeast TRRAP proteins (Saleh et al. 1998).
The paradox of sequence conservation in the absence of
kinase activity prompted us to explore a noncatalytic
role for this conserved domain in the recruitment of
HAT complexes to transcriptional activation domains.
Results
To investigate the function of the ATM-related region of
TRRAP, a progressive series of C-terminal deletions (1–
3760, 1–3713, and 1–3087) was constructed from a FLAG
epitope-tagged full-length (1–3830) TRRAP cDNA ex-
pression vector (Fig. 1A). An internal deletion (2108–
2403) within the TRRAP cDNA was also constructed.
Each protein was transiently expressed in HEK293 cells,
immunoprecipitated with anti-FLAG antibodies, eluted
from the beads with an excess of FLAG peptide, and sub-
jected to histone acetyltransferase assays using core his-
tones as substrates. The reaction products were resolved
by graded porosity SDS-PAGE and transferred onto ni-
trocellulose membranes. The upper section of the mem-
brane was probed with antibodies for FLAG and hGCN5
and the bottom section was subjected to fluorography
after Ponceau-S staining (Fig. 1B). We found that the full-
length TRRAP protein bound to the endogenous hGCN5
protein and that the immunoprecipitates had readily as-
sayed histone acetyltransferase activity. In contrast, all
of the C-terminal deletion mutations abolished TRRAP
binding to hGCN5 and the recruitment of histone acet-
yltransferase activity, including the TRRAP(1–3760) pro-
tein with only a 70-amino-acid deletion including the
FATC domain. In addition, an internal 20-amino-acid
deletion (3692–3713) of a highly conserved sequence
within the ATM-related domain also abolished the in-
teraction with hGCN5 (data not shown). An internal de-
letion (2108–2403), distant from the ATM-related do-
main, showed little or no difference in hGCN5 binding
or HAT activity compared with full-length TRRAP.
These data establish that the ATM-related domain of
TRRAP is necessary for HAT recruitment. To determine
whether the ATM-related domain of TRRAP was suffi-
cient for HAT recruitment, the C terminus of TRRAP
(3404–3830) was expressed with a FLAG epitope tag (Fig.
1B, lane 7). The smaller C-terminal domain was ex-
pressed at a much higher level than full-length TRRAP,
[Key Words: TRRAP; histone acetyltransferase; Myc; ATM domain; on-
cogenesis]
3
Corresponding author.
E-MAIL mcole@molbio.princeton.edu; FAX (609) 258-4575.
Article and publication are at http://www.genesdev.org/cgi/doi/10.1101/
gad.900101.
GENES & DEVELOPMENT 15:1619–1624 © 2001 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/01 $5.00; www.genesdev.org 1619
but this segment was inactive in binding to hGCN5 or in
the recruitment of histone acetyltransferase activity.
Thus, the ATM/PI-3 kinase homology domain is neces-
sary but not sufficient for the assembly of a functional
HAT enzyme complex with TRRAP.
The recruitment of hGCN5 by TRRAP is expected to
create an enzyme complex with predominantly H3-spe-
cific HAT activity (Yang et al. 1996; Wang et al. 1997),
but the TRRAP immunoprecipitates showed an equal
preference for H3 and H4 acetylation. The identity of the
H4-specific histone acetyltransferase in association with
TRRAP requires further characterization but could de-
rive from human MYST family proteins TIP60, MOZ, or
MORF (Sterner and Berger 2000).
Functional TRRAP and hGCN5 proteins are required
for cellular transformation by the c-Myc oncoprotein
(McMahon et al. 1998, 2000). The involvement of the
TRRAP ATM-related homology domain in the recruit-
ment of histone acetyltransferase activities prompted us
to investigate its physiological role in the cell cycle and
cellular transformation. Expression vectors for full-
length TRRAP or the TRRAP(1–3760) truncation mutant
were cotransfected with expression vectors for c-Myc
and/or oncogenic H-rasG12V into early-passage rat em-
bryo fibroblasts (Fig. 2A). Neither TRRAP itself nor the
truncation mutant had intrinsic transforming activity in
conjunction with the H-rasG12V oncogene. Cotransfec-
tion of c-myc and H-rasG12V leads to focus formation,
and the addition of the wild-type TRRAP expression vec-
Figure 2. Expression of a TRRAP mutant defective in HAT
recruitment inhibits c-Myc oncogenic activity. (A) Primary rat
embryo fibroblasts were transfected with expression vectors for
c-Myc, H-RasG12V, wild-type full-length TRRAP, and the
HAT-defective TRRAP(1–3760) truncation mutant in the differ-
ent combinations indicated. (yaxis) Number of transformed
foci. Three plates were assayed for each bar. (B) TRRAP and
TRRAP mutants do not inhibit colony growth. Primary rat em-
bryo fibroblasts were transfected as indicated along with pSG5
puromycin
R
plasmid. Puromycin
R
colonies were counted after
14 d. Two plates were assayed for each bar in the graph.
Figure 1. The ATM/PI-3 kinase domain of TRRAP is critical
for the recruitment of histone acetyltransferase activity. (A)
Schematic representation of wild-type, full-length TRRAP (top)
with the 300 C-terminal amino acids homologous to the ATM/
PI-3 kinase family members indicated as ATM. Below is shown
a series of TRRAP internal fragments, C-terminal truncation
mutants, and internal deletion mutants with their respective
sizes at the right. (B) ATM-domain dependence for HAT recruit-
ment. Complexes containing transiently expressed FLAG-
tagged TRRAP or TRRAP mutants were isolated from HEK293
cells (McMahon et al. 1998). The FLAG–TRRAP or TRRAP mu-
tants were eluted from beads and assayed for HAT activity on
calf thymus core histones. Reaction mixtures were resolved on
an SDS-PAGE gradient gel (4%–15%) and transferred to a nitro-
cellulose membrane. The upper section was probed with anti-
FLAG and anti-hGCN5 antibodies. Cell lysate containing 100
µg of protein was included as a positive control for hGCN5 (lane
left of lane 1). The bottom portion of the membrane below the
30-kD molecular mass marker was stained with Ponceau-S and
subjected to fluorography for HAT activities. The 97-kD band
common to lanes 17in the hGCN5 panel is a nonspecific band
that arises from the anti-FLAG beads, but is not found with cell
lysates (lane left of lane 1).
Parketal.
1620 GENES & DEVELOPMENT
tor had no effect on this activity. In contrast, cotransfec-
tion of c-myc and H-rasG12V with the TRRAP(1–3760)
mutant that is defective for HAT-recruitment resulted in
a 50% inhibition of focus formation. The finding of only
a partial dominant-negative phenotype with the
TRRAP(1–3760) mutant may be due to the large size of
the TRRAP protein and relatively low level of ectopic
expression, which may not allow effective competition
with the endogenous wild-type TRRAP. In parallel with
the REF transformation assay, we tested for transfection
efficiency or any toxicity from the TRRAP expression
constructs by scoring for puromycin-resistant colonies
after cotransfection with the pSG5-Puromycin
R
plasmid
(Fig. 2B). Approximately equal numbers of puromycin-
resistant colonies were obtained with all plasmid com-
binations tested, indicating that the inhibitory effect of
the TRRAP(1–3760) truncation mutant on oncogenic
transformation did not result from cellular toxicity or
general growth inhibition.
In addition to TRRAP–Tra1p and GCN5–PCAF, the
SAGA–PCAF complexes contain SPT3, which has been
suggested to regulate transcription through interaction
with TBP (Dudley et al. 1999; Belotserkovskaya et al.
2000). SPT3 protein has been found in all TRRAP com-
plexes that contain hGCN5 (Brand et al. 1999). Because
mutations in the ATM-homology region abolished bind-
ing to hGCN5, we were interested in determining what
other components of the complex were dependent on
this domain. To assess whether hSPT3 binding to
TRRAP was dependent on the ATM-related domain, im-
munoprecipitates of FLAG-tagged full-length or a C-ter-
minal deletion of TRRAP were probed with anti-SPT3
antibodies (Fig. 3). Full-length TRRAP bound to hSPT3,
but binding was abolished by a small deletion in the
ATM-related domain.
The exclusive enrichment of TRRAP by the c-Myc N
terminus in the initial affinity purification strategy sug-
gested that the interaction between these two nuclear
proteins is direct and unlikely to require any associated
cofactors (McMahon et al. 1998). We were interested in
determining which domain or motif in TRRAP is impor-
tant for c-Myc binding. Progressive deletions and several
internal fragments in the TRRAP C terminus were used
to map the sequence for Myc binding (Fig. 4). Plasmids
expressing FLAG-tagged TRRAP (or mutants thereof)
were transiently cotransfected into HEK293 cells with
an expression vector for c-Myc, and then anti-FLAG im-
munoprecipitates isolated under nondenaturing condi-
tions were probed by Western blot analysis for c-Myc
protein (Fig. 4A, bottom). The expression levels and re-
covery of the TRRAP proteins were tested by anti-FLAG
Western blotting (Fig. 4A, top), showing that smaller
TRRAP mutants were generally expressed better than
the full-length protein. In the initial experiments, dele-
tions from the C terminus up to amino acid 3087 re-
tained the ability to coprecipitate with c-Myc, in con-
trast with the hGCN5 binding described above. Expres-
sion of only the N-terminal half of TRRAP, 1–1700,
coprecipitated a negligible amount of c-Myc despite the
fact that the expression level was significantly higher
than the larger TRRAP proteins. These data indicate that
the Myc-binding site maps between amino acids 1700
and 3087. To refine the mapping further, three FLAG-
tagged internal fragments of TRRAP (1899–2401, 2402–
2869, and 2869–3470) were transiently expressed, immu-
noprecipitated with anti-FLAG antibody, and assayed for
binding to endogenous c-Myc by Western blot (Fig. 4B,
top). In parallel, immunoprecipitates by anti-FLAG anti-
bodies were analyzed by Western blotting to assess the
expression level of FLAG-tagged TRRAP fragments (Fig.
4B, bottom). Only the TRRAP(1899–2401) fragment co-
precipitates with endogenous c-Myc, suggesting that it
contains the primary c-Myc interaction domain in the
context of intact TRRAP.
Coprecipitation of c-Myc and TRRAP from whole cell
lysates could be mediated by another component of
SAGA or other intermediate protein. A more direct assay
of protein–protein interactions using the GST-pulldown
technique was used to map the Myc-binding domain on
TRRAP (Fig. 4C). To broaden the analysis, we tested the
binding of N-Myc to TRRAP as N-Myc binds with an
affinity equal to that of c-Myc (Wood et al. 2000). A se-
ries of TRRAP protein fragments was expressed to equal
levels in Escherichia coli and purified on beads. The
beads were then incubated with affinity-purified N-Myc
protein to assess binding. Five segments spanning 50%
of the 3830-amino-acid TRRAP protein were tested for
N-Myc binding, and only one segment (1591–2026) ex-
hibited significant binding compared with GST alone.
This segment overlaps the binding domain predicted
from mammalian expression studies above, suggesting
that the major site of Myc interaction with TRRAP maps
between amino acids 1899 and 2026.
To provide an in vivo test of the N-Myc–TRRAP in-
teraction domain, two FLAG–TRRAP protein segments
(1261–1579 and 1899–2401) were stably expressed at
equal levels in human neuroblastoma cells, which con-
tain an amplified N-myc gene (data not shown). Because
Myc is required for cell cycle progression, competition
between endogenous N-Myc and a critical cofactor
would be expected to impede cell growth. Consistent
with this model, expression of FLAG–TRRAP(1899–
2401) containing the Myc-binding domain increased the
Figure 3. The ATM/PI-3 kinase domain of TRRAP is critical
for interaction with hSPT3. HEK293 cells were transiently
transfected with expression vectors for FLAG-tagged TRRAP
fragment or its deletion mutant, TRRAP(1–3713). Cell lysates
were prepared and immunoprecipitated with anti-FLAG anti-
bodies. Each precipitate was resolved by SDS-PAGE and ana-
lyzed for the extent of its coprecipitation with endogenous
hSPT3. To check the relative expression levels of TRRAP frag-
ments and hGCN5 recruitment, the same membrane was
reprobed with anti-FLAG antibodies (top) and anti-hGCN5
(middle).
TRRAP interaction with HAT and Myc
GENES & DEVELOPMENT 1621
doubling time of IMR5 cells from 28 to 63 h, or 2.25-fold
(Table 1). Expression of another segment of TRRAP
(1261–1579) had no impact on growth rate compared
with control cells, even though the latter can inhibit
c-Myc and N-Myc transforming activity in primary fi-
broblasts (McMahon et al. 1998).
Discussion
This study provides the first functional mapping of do-
mains within the large ATM-related TRRAP protein that
is implicated in transcriptional regulation and chroma-
tin modification from yeast to man. We find that the
c-Myc-binding region is distinct from the ATM/PI-3 ki-
nase domain, and this region is conserved among TRRAP
orthologs from other species, but not conserved in other
ATM family members. Interestingly, this region over-
laps the direct binding site for transcriptional activators
on the yeast ortholog of TRRAP (Tra1p; J. Workman,
pers. comm.), suggesting that the function of TRRAP–
Tra1p as a mediator of HAT-activator interactions has
been highly conserved in evolution. Furthermore, this
same domain of Tra1p binds to Yng2p in yeast HAT
complexes and hence may play a major functional role in
all TRRAP–Tra1p complexes (Loewith et al. 2000).
Although the ATM-related C-terminal domain of
TRRAP lacks kinase activity, we show here that this
domain is essential for the recruitment of HAT activity
by the complex, and similar findings have been made for
the yeast Tra1p (J. Workman, pers. comm.). Generally
protein–protein interactions are mediated by relatively
small motifs, and, therefore, it was unexpected that mu-
tations in the ATM-related domain abolished HAT re-
cruitment whereas the isolated ATM-related domain is
defective for binding. These results suggest that TRRAP
may adopt a complex three-dimensional structure that is
dependent on the ATM homology domain for the re-
cruitment of HATs and most other components of the
complex. A parallel can be drawn with the ATM-related
Rad3p in fission yeast, which requires an N-terminal
protein domain for the activity of the C-terminal kinase
domain (Chapman et al. 1999). ATM/PI3 kinase family
proteins are frequently involved in checkpoint or sensing
functions within the cell, suggesting that the activity of
TRRAP itself may also be regulated.
Figure 4. TRRAP amino acids 1899–2026 mediate Myc bind-
ing. (A) In vivo coimmunoprecipitation. HEK293 cells were
transiently cotransfected with CMV-driven expression vectors
for c-Myc and FLAG-tagged full-length TRRAP or a series of
TRRAP C-terminal truncation mutants. Cell lysates were pre-
pared and the FLAG-tagged TRRAP protein or mutants were
immunoprecipitated with anti-FLAG antibodies. Precipitates
were resolved by SDS-PAGE and analyzed by Western blotting
for anti-c-Myc antibodies (bottom). The same membrane was
also probed with anti-FLAG antibodies to detect TRRAP expres-
sion (top). Apparent molecular masses in kilodaltons of protein
markers are indicated at the left of fluorogram. (B) Localization
of the c-Myc-binding domain. HEK 293 cells were transiently
transfected with expression vectors for FLAG-tagged TRRAP
fragment. Cell lysates were prepared and immunoprecipitated
with anti-FLAG antibodies. Each precipitate was resolved by
SDS-PAGE and analyzed for the extent of its coprecipitation
with endogenous c-Myc (top). Cell lysate from the c-Myc trans-
fection was included as a positive control (lane left of lane 1). To
check the relative expression levels of TRRAP fragments, the
same membrane was reprobed with anti-FLAG antibodies (bot-
tom). (C) An internal region of TRRAP (spanning amino acids
1591–2026) interacts with N-Myc. GST fusion proteins contain-
ing the indicated TRRAP regions were expressed in E. coli and
bound to glutathione beads. Equal amounts of bead-bound fu-
sion proteins were incubated with affinity-purified FLAG-
tagged N-Myc protein. After washing, interacting proteins were
subjected to SDS-PAGE analysis and immunoblotted with anti-
FLAG antibody. The FLAG-N-Myc starting material (20% of
input) was run with the bound material.
Table 1. Myc-interacting region of TRRAP has a dominant
negative effect on cell growth
Cell line TRRAP segment Doubling time (h)
IMR-5 26.7 ± 6.4
IMR-5 1261–1579 27.9 ± 5.3
IMR-5 1899–2401 63.1 ± 12.2
IMR-5 human neuroblastoma cells were stably transfected with
expression vectors for FLAG-tagged TRRAP fragments as indi-
cated. Cell lysates were immunoprecipitated with anti-FLAG
antibody followed by immunoblot analysis with the same anti-
body (data not shown). Both fragments were comparably ex-
pressed. Doubling times were calculated from three individual
plates from each line SE).
Parketal.
1622 GENES & DEVELOPMENT
A common mechanism of chromatin modification is
the acetylation of N-terminal lysine residues on his-
tones, which reduces histone–DNA interactions to pro-
mote a more open chromatin configuration (Grant et al.
1997). A key feature of the multiprotein HAT complexes
is the ability to acetylate nucleosomal histones, whereas
the HAT enzyme alone can only acetylate free histones
(Kuo et al. 1996; Grant et al. 1997). Therefore, one or
more components of these complexes must modify HAT
activity to facilitate nucleosome recognition, as well as
mediate binding to transcriptional activators. TRRAP is
remarkably conserved in evolution as a 3800 ± 100-
amino-acid polypeptide, implying that size and the ori-
entation of functional elements are critical. TRRAP–
Tra1p appears to be a direct, global mediator of transcrip-
tional activator–HAT interactions, and further
elucidation of its structure and function may provide
important insights into the dynamics of transcriptional
activation and chromatin modification.
Materials and methods
Plasmid constructs and transfections
Expression plasmids were created by standard methods in a CMV-pro-
moter driven vector containing a FLAG-epitope tag and verified by se-
quence analysis (McMahon et al. 1998). Details of individual constructs
are available on request.
HAT assays
FLAG-tagged TRRAP protein was transiently expressed in HEK293 cells
and isolated from whole-cell lysates by binding to anti-FLAG antibody
covalently conjugated to beads (Sigma). The complexes containing
FLAG–TRRAP or its mutants were released by addition of excess FLAG
peptide in HAT assay buffer (50 mM Tris at pH 8.0, 10% glycerol, 50 mM
KCl, 0.1 mM EDTA, 10 mM butyric acid, 1 mM DTT, 1 mM PMSF). Calf
thymus core histones (3 µg) along with
14
C-acetyl-CoA were incubated
with each eluate, and reaction mixtures were resolved on a SDS-PAGE
gradient gel (4%–15%; Ogryzko et al. 1996). Proteins were transfered
onto a nitrocellulose membrane, and the section above the 30-kD mo-
lecular mass marker was subjected to Western blot analysis and probed
with anti-FLAG and anti-hGCN5 antibodies. The bottom section of the
membrane was stained with Ponceau-S and subjected to fluorography for
HAT activities.
GST pulldown assay
TRRAP fragments were PCR amplified, cloned into pGXT4T1 (Amer-
sham Pharmacia Biotech) and expressed as fusion proteins in E. coli. For
large-scale purification, bacterial pellets were sonicated in STE buffer (10
mM Tris at pH 8.0, 150 mM NaCl, 1 mM EDTA, 5 mM DTT, 1.5%
Sarkosyl, PMSF–aprotinin). After addition of Triton X-100 to a final con-
centration of 2%, lysates were cleared by centrifugation, and the fusion
proteins were bound to glutathione–Sepharose 4B beads (Amersham
Pharmacia Biotech). Beads were analyzed for the fusion protein by SDS-
PAGE followed by Western analysis for GST.
FLAG–N-Myc was stably expressed from a CMV promoter in IMR-5
neuroblastoma cells after neo cotransfection and G418 selection. Cells
from three semiconfluent 15-cm plates were lysed in E1A lysis buffer
(McMahon et al. 1998), followed by overnight immunoprecipitation with
anti-FLAG antibody (Sigma) and Protein A/G beads (Santa Cruz). Bound
FLAG–N-Myc was eluted with FLAG peptide (Sigma). Equal amounts of
GST–TRRAP or control beads (based on the Western), were incubated
with 30–50 µL of the FLAG–N-Myc proteins in binding buffer for 3 h in
4°C. Beads were washed three times in the same buffer, and then bound
proteins were resolved by SDS-PAGE. N-Myc interaction was detected
with anti-FLAG antibody, and the same blot was reprobed with anti-GST
to assess the amount of protein on the beads.
Cell culture
Rat embryo fibroblasts were prepared by trypsinization of 15-d embryos
(Land 1995), and focus formation was assessed as described (McMahon et
al. 1998). Foci were scored after 18 d. Growth properties after transfection
of the same DNAs were assessed on duplicate plates of primary rat em-
bryo fibroblasts cotransfected as indicated along with 1 µg of pSG5 pu-
romycin
R
plasmid. Transfected cells were selected in 2.5 µg/mL puro-
mycin for 14 d, at which time the number of colonies per plate was
determined.
FLAG–TRRAP fragments were stably expressed in IMR-5 neuroblas-
toma cells by cotransfection with neo and selection with G418. Growth
rates and protein expression were assessed at early passage of G418-
resistant polyclonal populations. Equal numbers of viable cells were
plated in parallel for the parental IMR-5 line and the two TRRAP frag-
ment-expressing lines. Cells were counted 48 and 72 h after plating, and
the doubling time was calculated.
Acknowledgments
We thank John Maris for providing IMR-5 cells, and Christine Brown and
Jerry Workman for communicating results in advance of publication.
This work was supported by a grant from the National Cancer Institute
to MDC. This work was also supported by a Leukemia Society Special
Fellow award and grants from the Edward Mallinckrodt, Jr. Foundation,
the Mary L. Smith Charitable Lead Trust, the Gustavus and Louise
Pfeiffer Research Foundation, and the American Association for Cancer
Research to S.B.M.
The publication costs of this article were defrayed in part by payment
of page charges. This article must therefore be hereby marked “adver-
tisement” in accordance with 18 USC section 1734 solely to indicate this
fact.
References
Belotserkovskaya, R., Sterner, D.E., Deng, M., Sayre, M.H., Lieberman,
P.M., and Berger, S.L. 2000. Inhibition of TATA-binding protein func-
tion by SAGA subunits Spt3 and Spt8 at Gcn4-activated promoters.
Mol. Cell. Biol. 20: 634–647.
Bosotti, R., Isacchi, A., and Sonnhammer, E.L. 2000. FAT: A novel do-
main in PIK-related kinases. Trends Biochem. Sci. 25: 225–227.
Brand, M., Yamamoto, K., Staub, A., and Tora, L. 1999. Identification of
TATA-binding protein-freeTAFII-containing complex subunits sug-
gests a role in nucleosome acetylation and signal transduction. J.
Biol. Chem. 274: 18285–18289.
Chapman, C.R., Evans, S.T., Carr, A.M., and Enoch, T. 1999. Require-
ment of sequences outside the conserved kinase domain of fission
yeast Rad3p for checkpoint control. Mol. Biol. Cell 10: 3223–3238.
Dudley, A.M., Rougeulle, C., and Winston, F. 1999. The Spt components
of SAGA facilitate TBP binding to a promoter at a post-activator-
binding step in vivo. Genes &Dev. 13: 2940–2945.
Grant, P.A., Duggan, L., Cote, J., Roberts, S.M., Brownell, J.E., Candau,
R., Ohba, R., Owen-Hughes, T., Allis, C.D., Winston, F., et al. 1997.
Yeast Gcn5 functions in two multisubunit complexes to acetylate
nucleosomal histones: Characterization of an Ada complex and the
SAGA (Spt/Ada) complex. Genes &Dev. 11: 1640–1650.
Grant, P.A., Schieltz, D., Pray-Grant, M.G., Yates, J.R.R., and Workman,
J.L. 1998. The ATM-related cofactor Tra1 is a component of the pu-
rified SAGA complex. Mol. Cell 2: 863–867.
Keith, C.T. and Schreiber, S.L. 1995. PIK-related kinases: DNA repair,
recombination, and cell cycle checkpoints. Science 270: 50–51.
Kuo, M.-H., Brownell, J.E., Sobel, R.E., Ranalli, T.A., Cook, R.G., Edmon-
son, D.G., Roth, S.Y., and Allis, C.D. 1996. GCN5p, a yeast nuclear
histone acetyltransferase, acetylates specific lysines in histones H3
and H4 that differ from deposition-related acetylation sites. Nature
383: 269–272.
Land, H. 1995. Transformation of primary rat embryo cells. Methods
Enzymol. 254: 37–41.
Loewith, R., Meijer, M., Lees-Miller, S.P., Riabowol, K., and Young, D.
2000. Three yeast proteins related to the human candidate tumor
suppressor p33(ING1) are associated with histone acetyltransferase
activities. Mol. Cell. Biol. 20: 3807–3816.
McMahon, S.B., Van Buskirk, H.A., Dugan, K.A., Copeland, T.D., and
Cole, M.D. 1998. The novel ATM-related protein TRRAP is an es-
sential cofactor for the c-Myc and E2F oncoproteins. Cell 94: 363–
374.
McMahon, S.B., Wood, M.A., and Cole, M.D. 2000. The c-Myc cofactor
TRRAP interaction with HAT and Myc
GENES & DEVELOPMENT 1623
TRRAP recruits the histone acetylase hGCN5. Mol. Cell. Biol. 20:
556–562.
Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H., and Nakatani,
Y. 1996. The transcriptional coactivators p300 and CBP are histone
acetyltransferases. Cell 87: 953–959.
Saleh, A., Schieltz, D., Ting, N., McMahon, S.B., Litchfield, D.W., Yates
III, J.R., Lees-Miller, S.P., Cole, M.D., and Brandl, C.J. 1998. Tra1p is
a component of the yeast ADA/SPT transcriptional regulatory com-
plexes. J. Biol. Chem. 273: 26559–26570.
Sterner, D.E. and Berger, S.L. 2000. Acetylation of histones and transcrip-
tion-related factors. Microbiol. Mol. Biol. Rev. 64: 435–459.
Vassilev, A., Yamauchi, J., Kotani, T., Prives, C., Avantaggiati, M.L., Qin,
J., and Nakatani, Y. 1998. The 400 kDa subunit of the PCAF histone
acetylase complex belongs to the ATM superfamily. Mol. Cell 2:
869–875.
Wang, L., Mizzen, C., Ying, R., Candau, R., Barlev, N., Brownell, J., Allis,
C.D., and Berger, S. 1997. Histone acetyltransferase activity is con-
served between yeast and human GCN5 and required for comple-
mentation of growth and transcriptional activation. Mol. Cell. Biol.
17: 519–527.
Wood, M.A., McMahon, S.B., and Cole, M.D. 2000. An ATPase/helicase
complex is an essential cofactor for oncogenic transformation by c-
Myc. Mol. Cell 5: 321–330.
Yang, X.-J., Ogryzko, V.V., Nishikawa, J., Howard, B.H., and Nakatani, Y.
1996. A p300/CBP-associated factor that competes with the adeno-
viral oncoprotein E1A. Nature 382: 319–324.
Parketal.
1624 GENES & DEVELOPMENT
... To test the genetic requirement of the core module, we depleted the SAGA-specific core subunit WDA by generating germ line clones of wda [11] (Fig 1C). Since the wda [11] allele also disrupts a neighboring gene [42], we performed an ethyl methanesulfonate (EMS) mutagenesis screen to isolate a new mutant allele of wda (wda[EMS]) ( Fig 1D). ...
... To test the genetic requirement of the core module, we depleted the SAGA-specific core subunit WDA by generating germ line clones of wda [11] (Fig 1C). Since the wda [11] allele also disrupts a neighboring gene [42], we performed an ethyl methanesulfonate (EMS) mutagenesis screen to isolate a new mutant allele of wda (wda[EMS]) ( Fig 1D). In ovaries with wda [11] or wda[EMS] germ line clones, the number of round early-stage egg chambers increased and latestage egg chambers did not form, indicating a failure to progress through stage 7 of mid-oogenesis. ...
... Since the wda [11] allele also disrupts a neighboring gene [42], we performed an ethyl methanesulfonate (EMS) mutagenesis screen to isolate a new mutant allele of wda (wda[EMS]) ( Fig 1D). In ovaries with wda [11] or wda[EMS] germ line clones, the number of round early-stage egg chambers increased and latestage egg chambers did not form, indicating a failure to progress through stage 7 of mid-oogenesis. ...
Preprint
Full-text available
The Spt/Ada-Gcn5 Acetyltransferase (SAGA) coactivator complex has multiple modules with different enzymatic and non-enzymatic functions. How each module contributes to gene activation in specific biological contexts is not well understood. Here we analyzed the role of the non-enzymatic core module during Drosophila oogenesis. We show that depletion of several SAGA-specific subunits belonging to the core module blocked egg chamber development during mid-oogenesis stages, resulting in stronger phenotypes than those obtained after depletion of SAGA’s histone acetyltransferase module or deubiquitination module. These results, as well as additional genetic analyses pointing to an interaction with TBP, suggested a differential role of SAGA modules at different promoter types. However, SAGA subunits co-occupied all promoter types of active genes in ChIP-seq and ChIP-nexus experiments. Thus, the SAGA complex appears to occupy promoters in its entirety, consistent with the strong biochemical integrity of the complex. The high-resolution genomic binding profiles are congruent with SAGA recruitment by activators upstream of the start site, and retention on chromatin by interactions with modified histones downstream of the start site. The stronger genetic requirement of the core module during oogenesis may therefore be explained through its interaction with TBP or its role in recruiting the enzymatic modules to the promoter. We propose the handyman principle, which posits that a distinct genetic requirement for specific components may conceal the fact that the entire complex is physically present. Author Summary Embryonic development critically relies on the differential expression of genes in different tissues. This involves the dynamic interplay between DNA, sequence-specific transcription factors, coactivators and chromatin remodelers which guide the transcription machinery to the appropriate promoters for productive transcription. To understand how this happens at the molecular level, we need to understand when and how coactivator complexes such as SAGA function. SAGA consists of multiple modules with well characterized enzymatic functions. This study shows that the non-enzymatic core module of SAGA is required for Drosophila oogenesis, while the enzymatic functions are largely dispensable. Despite this differential requirement, SAGA subunits appear to be broadly recruited to all promoter types, consistent with the biochemical integrity of the complex. These results suggest that genetic requirements and physical organization do not always go hand in hand.
... TRRAP was initially discovered as a coactivator for the c-MYC and E2F transcription factors and is essential for their oncogenic activities . Further work demonstrated that many additional activators require Tra1 or TRRAP to stimulate transcription initiation (Park et al., 2001;Bouchard et al., 2001;Lang et al., 2001;Deleu et al., 2001;Ard et al., 2002;Lang and Hearing, 2003;Memedula and Belmont, 2003;Knutson and Hahn, 2011;Lin et al., 2012). Work in yeast demonstrated that Tra1 physically interacts with the transactivation domain of activators in vivo (Brown et al., 2001;Bhaumik and Green, 2001;Bhaumik et al., 2004;Fishburn et al., 2005;Reeves and Hahn, 2005). ...
... Notably, we found that TELO2 is important for the activation of MYC and E2F target genes (Figure 2). In agreement, TRRAP is essential for transcription activation by MYC and E2F and contributes to their oncogenic functions during tumorigenesis Park et al., 2001;Bouchard et al., 2001;Lang et al., 2001;Nikiforov et al., 2002). Unexpectedly, we found that TELO2 and TRRAP also repress several genes, particularly those mediating the IFN-I response during innate immunity. ...
Article
Full-text available
Transcription is essential for cells to respond to signaling cues and involves factors with multiple distinct activities. One such factor, TRRAP, functions as part of two large complexes, SAGA and TIP60, which have crucial roles during transcription activation. Structurally, TRRAP belongs to the PIKK family but is the only member classified as a pseudokinase. Recent studies established that a dedicated HSP90 co-chaperone, the TTT complex, is essential for PIKK stabilization and activity. Here, using endogenous auxin-inducible degron alleles, we show that the TTT subunit TELO2 promotes TRRAP assembly into SAGA and TIP60 in human colorectal cancer cells (CRC). Transcriptomic analysis revealed that TELO2 contributes to TRRAP regulatory roles in CRC cells, most notably of MYC target genes. Surprisingly, TELO2 and TRRAP depletion also induced the expression of type I interferon genes. Using a combination of nascent RNA, antibody-targeted chromatin profiling (CUT&RUN), ChIP, and kinetic analyses, we propose a model by which TRRAP directly represses the transcription of IRF9, which encodes a master regulator of interferon stimulated genes. We have therefore uncovered an unexpected transcriptional repressor role for TRRAP, which we propose contributes to its tumorigenic activity.
... Activation involves the recruitment of multiple coactivators and protein complexes to E-box elements. The TIP60 acetyltransferase complex and the histone acetyltransferase GCN5 are bound to MYC indirectly through the TRRAP adaptor protein that interacts with MBII of the MYC protein (67)(68)(69)(70). Two other proteins, TIP48 and TIP49, found in the TIP60 complex, are involved in chromatin remodeling and bind to the N-terminus of MYCN (67). ...
... MYCN recruits many HDACs (HDAC1, HDAC2 and HDAC5) to repress gene transcription (74,135,136). The histone acetyltransferase, GCN5, binds to MYC and MYCN proteins (67)(68)(69). In vitro luciferase assays show that MYC recruits GCN5 to activate gene transcription (70); however, few GCN5 specific inhibitors are available and have limited testing in NB cells (137). ...
Article
Full-text available
The deregulation of the MYC family of oncogenes, including c-MYC, MYCN and MYCL occurs in many types of cancers, and is frequently associated with a poor prognosis. The majority of functional studies have focused on c-MYC due to its broad expression profile in human cancers. The existence of highly conserved functional domains between MYCN and c-MYC suggests that MYCN participates in similar activities. MYC encodes a basic helix-loop-helix-leucine zipper (bHLH-LZ) transcription factor (TF) whose central oncogenic role in many human cancers makes it a highly desirable therapeutic target. Historically, as a TF, MYC has been regarded as “undruggable”. Thus, recent efforts focus on investigating methods to indirectly target MYC to achieve anti-tumor effects. This review will primarily summarize the recent progress in understanding the function of MYCN. It will explore efforts at targeting MYCN, including strategies aimed at suppression of MYCN transcription, destabilization of MYCN protein, inhibition of MYCN transcriptional activity, repression of MYCN targets and utilization of MYCN overexpression dependent synthetic lethality.
... A module is defined as a complex of two protein molecules (Poluri et al. 2021e). For example, Youtiao, a scaffolding protein, binds with NMDA receptor and potassium ion channels and facilitates their binding to protein kinase A and protein phosphatase 1, respectively (Marx et al. 2002;Park et al. 2001). Until now, a wide array of techniques have been employed to understand PPIs. ...
Chapter
Anomalies in protein–protein interactions may lead to a diseased state, as many cellular functions are sustained by such interactions. These abnormal situations usually arise when one or more of the interacting partners are mutated to an extent such that the original interaction is lost, or affinity toward a new interacting partner is developed. This chapter delineates the various human diseases that arise from aberrant protein interactions. It also discusses some of the experimental and computational methods that have been employed to determine such anomalous interactions as the underlying cause of plethora of diseases.
... This complex process generates high-affinity B-cell receptors through somatic hypermutation and selection. MYC translocation drives the expansion of BL cells within the germinal center, promoting their survival and proliferation, facilitated by the interaction with the microenvironment, including T-cells and stromal cells [84] Epigenetic regulation of MYC in BL expression and progression has also been the focus of many studies [85][86][87][88]. Fernández-Serrano et al. [89] reviewed the role of epigenetic modifications, such as histone modifications H3K4me3 and H3K27ac, in regulating MYC expression in BL cells. ...
Article
Full-text available
MYC deregulation, a cardinal event in Burkitt lymphoma (BL) pathogenesis, necessitates the elucidation of the molecular mechanisms governing MYC activation to devise innovative and effective therapeutic strategies. The t(8;14)(q24;q32) chromosomal translocation commonly observed in hematological malignancies results in MYC deregulation, endowing cancer cells with a competitive edge through heightened cell proliferation, cell cycle progression, apoptosis evasion, and metabolic reprogramming. Recent discoveries of recurrent MYC mutations in BL underscore the potential of precision medicine, employing tailored therapeutics to specifically inhibit MYC activity. However, the intricate genetic landscape of BL, featuring additional alterations, such as mutations in TP53, TCF3, and ID3, may necessitate a combinatorial approach targeting multiple oncogenic pathways for effective intervention. Despite significant strides in hematological malignancy treatment, a comprehensive understanding of the molecular mechanisms underpinning MYC’s oncogenic properties remains crucial for the potential development of highly potent and selective MYC-directed cancer therapies. This review offers an in-depth analysis of MYC translocation and its implications in Burkitt lymphoma, with a spotlight on cutting-edge advances in research and emerging therapeutic paradigms.
... TRRAP mutation which was identified in BRAF wildtype PTC patients (61), plays an important role in the recruitment of histone acetyltransferase (HAT) complexes to the chromatin, regulating transcription and DNA repair. Its FATC domain can bind to Myc, regulating Myc oncogenic activities (62,63). In this present study, the TRRAP p.S2912T mutation is located within the FAT domain, where the function and protein-protein interaction of this domain remains largely unknown. ...
Article
Full-text available
Background Papillary thyroid cancer (PTC) is the most common thyroid malignancy. Concurrent presence of cytomorphological benign thyroid goitre (BTG) and PTC lesion is often detected. Aberrant protein profiles were previously reported in patients with and without BTG cytomorphological background. This study aimed to evaluate gene mutation profiles to further understand the molecular mechanism underlying BTG, PTC without BTG background and PTC with BTG background. Methods Patients were grouped according to the histopathological examination results: (i) BTG patients (n = 9), (ii) PTC patients without BTG background (PTCa, n = 8), and (iii) PTC patients with BTG background (PTCb, n = 5). Whole-exome sequencing (WES) was performed on genomic DNA extracted from thyroid tissue specimens. Nonsynonymous and splice-site variants with MAF of ≤ 1% in the 1000 Genomes Project were subjected to principal component analysis (PCA). PTC-specific SNVs were filtered against OncoKB and COSMIC while novel SNVs were screened through dbSNP and COSMIC databases. Functional impacts of the SNVs were predicted using PolyPhen-2 and SIFT. Protein-protein interaction (PPI) enrichment of the tumour-related genes was analysed using Metascape and MCODE algorithm. Results PCA plots showed distinctive SNV profiles among the three groups. OncoKB and COSMIC database screening identified 36 tumour-related genes including BRCA2 and FANCD2 in all groups. BRAF and 19 additional genes were found only in PTCa and PTCb. “Pathways in cancer”, “DNA repair” and “Fanconi anaemia pathway” were among the top networks shared by all groups. However, signalling pathways related to tyrosine kinases were the most significantly enriched in PTCa while “Jak-STAT signalling pathway” and “Notch signalling pathway” were the only significantly enriched in PTCb. Ten SNVs were PTC-specific of which two were novel; DCTN1 c.2786C>G (p.Ala929Gly) and TRRAP c.8735G>C (p.Ser2912Thr). Four out of the ten SNVs were unique to PTCa. Conclusion Distinctive gene mutation patterns detected in this study corroborated the previous protein profile findings. We hypothesised that the PTCa and PTCb subtypes differed in the underlying molecular mechanisms involving tyrosine kinase, Jak-STAT and Notch signalling pathways. The potential applications of the SNVs in differentiating the benign from the PTC subtypes requires further validation in a larger sample size.
... In connection, a meta-analysis of mRNA expression profile identified EP400NL being upregulated in lung adenocarcinoma tissue from cancer patients who have a smoking history [55]. Multiple transcription factors and epigenetic protein complexes including the hNuA4, EP400, and BRG1-containing BAF complexes have been identified as interacting partners with cMyc to induce and maintain cancerous phenotypes [2,7,8,34,[56][57][58][59][60][61][62][63]. Given that cMyc regulates ~15 % of all human genes, it is surprising to see only a small number of genes to be EP400NL responsive in our transcriptome analysis. ...
Article
Full-text available
EP400 is an ATP-dependent chromatin remodelling enzyme that regulates DNA double-strand break repair and transcription, including cMyc-dependent gene expression. We previously showed that the N-terminal domain of EP400 increases the efficacy of chemotherapeutic drugs against cancer cells. As the EP400 N-terminal-Like (EP400NL) gene resides next to the EP400 gene locus, this prompted us to investigate whether EP400NL plays a similar role in transcriptional regulation to the full-length EP400 protein. We found that EP400NL forms a human NuA4-like chromatin remodelling complex that lacks both the TIP60 histone acetyltransferase and EP400 ATPase. However, this EP400NL complex displays H2A.Z deposition activity on a chromatin template comparable to the human NuA4 complex, suggesting another associated ATPase such as BRG1 or RuvBL1/RuvBL2 catalyses the reaction. We demonstrated that the transcriptional coactivator function of EP400NL is required for serum and IFNγ-induced PD-L1 gene activation. Furthermore, transcriptome analysis indicates that EP400NL contributes to cMyc-responsive mitochondrial biogenesis. Taken together, our studies show that EP400NL plays a role as a transcription coactivator of PD-L1 gene regulation and provides a potential target to modulate cMyc functions in cancer therapy.
... TRRAP was shown to interact with MYC, more precisely with MBII (Buchel et al. 2017;Feris, Hinds, and Cole 2019;Frank et al. 2003;Kalkat et al. 2018;Liu et al. 2003;McMahon et al. 1998;McMahon, Wood, and Cole 2000;Nikiforov et al. 2002;Park et al. 2001;Park, Wood, and Cole 2002;Tu et al. 2015). Like this, MYC recruits HATs to chromatin via TRRAP to open up chromatin and facilitate transcription of its target genes (Bouchard et al. 2001;Frank et al. 2003;McMahon, Wood, and Cole 2000;Wood et al. 2000). ...
Thesis
Full-text available
The transcription factor MYC is a onco-protein, found to be deregulated in many human cancers. High MYC levels correlate with an aggressive tumor outcome and poor survival rates. Despite MYC being discovered as an oncogene already in the 1970s, how MYC regulates transcription of its target genes, which are involved in cellular growth and proliferation, is not fully understood yet. In this study, the question how MYC influences factors interacting with the RNA polymerase II ensuring productive transcription of its target genes was addressed using quantitative mass spectrometry. By comparing the interactome of RNA polymerase II under varying MYC levels, several potential factors involved in transcriptional elongation were identified. Furthermore, the question which of those factors interact with MYC was answered by employing quantitative mass spectrometry of MYC itself. Thereby, the direct interaction of MYC with the transcription elongation factor SPT5, a subunit of the DRB-sensitivity inducing factor, was discovered and analyzed in greater detail. SPT5 was shown to be recruited to chromatin by MYC. In addition, the interaction site of MYC on SPT5 was narrowed down to its evolutionary conserved NGN-domain, which is the known binding site for SPT4, the earlier characterized second subunit of the DRB-sensitivity inducing factor. This finding suggests a model in which MYC and SPT4 compete for binding the NGN-domain of SPT5. Investigations of the SPT5-interacting region on MYC showed binding of SPT5 to MYC’s N-terminus including MYC-boxes 0, I and II. In order to analyze proteins interacting specifically with the N-terminal region of MYC, a truncated MYC-mutant was used for quantitative mass spectrometric analysis uncovering reduced binding for several proteins including the well-known interactor TRRAP and TRRAP-associated complexes. Summarized, ...
... Moreover, a meta-analysis of mRNA expression profile identified EP400NL being upregulated in lung adenocarcinoma tissue from the cancer patients who have a smoking history (55). Multiple transcription factors and epigenetic protein complexes including the hNuA4, EP400, and BRG1-containing BAF complexes have been identified as interacting partners with Myc to induce and maintain cancerous phenotypes (2,7,8,29,(32)(33)(34)51,(56)(57)(58)(59). ...
Preprint
Full-text available
EP400 is an ATP-dependent chromatin remodeling enzyme that has been implicated in DNA double-strand break repair and transcription regulation including Myc-dependent gene expression. We previously showed that the N-terminal domain of EP400 increases the efficacy of chemotherapeutic drugs against cancer cells. As the EP400 N-terminal-Like (EP400NL) gene resides next to the EP400 gene locus prompted us to investigate whether EP400NL also plays a similar role in epigenetic transcriptional regulation to the full-length EP400 protein. We found that EP400NL forms a human NuA4-like chromatin remodelling complex that lacks both the TIP60 histone acetyltransferase and EP400 ATPase. However, this EP400NL complex displays H2A.Z deposition activity on a chromatin template comparable to the human NuA4 complex, suggesting another associated ATPase such as BRG1 or RuvBL1/RuvBL2 catalyses the reaction. We also demonstrated that the transcriptional coactivator function of EP400NL is required for cMyc and IFNγ-mediated PD-L1 gene activation. Collectively, our studies show that EP400NL plays a role as a transcription coactivator for cMyc-mediated gene expression and provides a potential target to modulate PD-L1 expression in cancer immunotherapy.
... Curcumin can form covalent thiol-Michael adducts, which could explain the observed cross-link between MYC and TRRAP complex on a molecular level. The MYC-Box II of human MYC contains one cysteine residue at position 133 and TRRAP has several cysteines in a region, which has been originally identified as MYCinteracting domain (18,60). It is assumed that sulfhydrylgroups of such cysteine residues form thiol adducts with the curcumin's enone electrophile of the heptadienone chain (21). ...
Article
Full-text available
The c-Myc protein (MYC) is a transcription factor with strong oncogenic potential controlling fundamental cellular processes. In most human tumors, MYC is overexpressed by enhanced transcriptional activation, gene amplification, chromosomal rearrangements, or increased protein stabilization. To pharmacologically suppress oncogenic MYC functions, multiple approaches have been applied either to inhibit transcriptional activation of the endogenous MYC gene, or to interfere with biochemical functions of aberrantly activated MYC. Other critical points of attack are targeted protein modification, or destabilization leading to a non-functional MYC oncoprotein. It has been claimed that the natural compound curcumin representing the principal curcumoid of turmeric (Curcuma longa) has anticancer properties although its specificity, efficacy, and the underlying molecular mechanisms have been controversially discussed. Here, we have tested curcumin’s effect on MYC-dependent cell transformation and transcriptional activation, and found that this natural compound interferes with both of these MYC activities. Furthermore, in curcumin-treated cells, the endogenous 60-kDa MYC protein is covalently and specifically cross-linked to one of its transcriptional interaction partners, namely the 434-kDa transformation/transcription domain associated protein (TRRAP). Thereby, endogenous MYC levels are strongly reduced and cells stop to proliferate. TRRAP is a multidomain adaptor protein of the phosphoinositide 3-kinase-related kinases (PIKK) family and represents an important component of many histone acetyltransferase (HAT) complexes. TRRAP is important to mediate transcriptional activation executed by the MYC oncoprotein, but on the other hand TRRAP also negatively regulates protein stability of the tumor suppressor p53 (TP53). Curcumin-mediated covalent binding of MYC to TRRAP reduces the protein amounts of both interaction partners but does not downregulate TP53, so that the growth-arresting effect of wild type TP53 could prevail. Our results elucidate a molecular mechanism of curcumin action that specifically and irreversibly targets two crucial multifunctional cellular players. With regard to their broad impact in cancer, our findings contribute to explain the pleiotropic functions of curcumin, and suggest that this natural spice, or more bioavailable derivatives thereof, may constitute useful adjuvants in the therapy of MYC-dependent and TRRAP-associated human tumors.
Article
Full-text available
SAGA is a 1.8-MDa yeast protein complex that is composed of several distinct classes of transcription-related factors, including the adaptor/acetyltransferase Gcn5, Spt proteins, and a subset of TBP-associated factors. Our results indicate that mutations that completely disrupt SAGA (deletions of SPT7 orSPT20) strongly reduce transcriptional activation at theHIS3 and TRP3 genes and that Gcn5 is required for normal HIS3 transcriptional start site selection. Surprisingly, mutations in Spt proteins involved in the SAGA-TBP interaction (Spt3 and Spt8) cause derepression of HIS3 andTRP3 transcription in the uninduced state. Consistent with this finding, wild-type SAGA inhibits TBP binding to theHIS3 promoter in vitro, while SAGA lacking Spt3 or Spt8 is not inhibitory. We detected two distinct forms of SAGA in cell extracts and, strikingly, one lacks Spt8. Conditions that induceHIS3 and TRP3 transcription result in an altered balance between these complexes strongly in favor of the form without Spt8. These results suggest that the composition of SAGA may be dynamic in vivo and may be regulated through dissociable inhibitory subunits.
Article
Full-text available
The adenoviral oncoprotein E1A induces progression through the cell cycle by binding to the products of the p300/CBP and retinoblastoma gene families. A new cellular p300/CBP-associated factor (P/CAF) having intrinsic histone acetylase activity has been identified that competes with E1A. Exogenous expression of P/CAF in HeLa cells inhibits cell-cycle progression and counteracts the mitogenic activity of E1A. E1A disturbs the normal cellular interaction between p300/CBP and its associated histone acetylase.
Article
Full-text available
Yeast and human ADA2 and GCN5 (y- and hADA2 and y- and hGCN5, respectively) have been shown to potentiate transcription in vivo and may function as adaptors to bridge physical interactions between DNA-bound activators and the basal transcriptional machinery. Recently it was shown that yGCN5 is a histone acetyltransferase (HAT), suggesting a link between enzymatic modification of nucleosomes and transcriptional activation. In this report, we demonstrate that hGCN5 is also an HAT and has the same substrate specificity as yGCN5. Since hGCN5 does not complement functional defects caused by deletion of yGCN5, we constructed a series of hGCN5-yGCN5 chimeras to identify human regions capable of activity in yeast. Interestingly, only the putative HAT domain of hGCN5, when fused to the remainder of yGCN5, complemented gcn5- cells for growth and transcriptional activation. Moreover, an amino acid substitution mutation within the HAT domain reduced both HAT activity in vitro and transcription in vivo. These findings directly link enzymatic histone acetylation and transcriptional activation and show evolutionary conservation of this potentially crucial pathway in gene regulation.
Article
Full-text available
The transcriptional adaptor protein Gcn5 has been identified as a nuclear histone acetyltransferase (HAT). Although recombinant yeast Gcn5 efficiently acetylates free histones, it fails to acetylate histones contained in nucleosomes, indicating that additional components are required for acetylation of chromosomal histones. We report here that Gcn5 functions as a catalytic subunit in two high-molecular-mass native HAT complexes, with apparent molecular masses of 0.8 and 1.8 megadalton (MD), respectively, which acetylate nucleosomal histones. Both the 0.8- and 1.8-MD Gcn5-containing complexes cofractionate with Ada2 and are lost in gcn5delta, ada2delta, or ada3delta yeast strains, illustrating that these HAT complexes are bona fide native Ada-transcriptional adaptor complexes. Importantly, the 1.8-MD adaptor/HAT complex also contains Spt gene products that are linked to TATA-binding protein (TBP) function. This complex is lost in spt20/ada5delta and spt7delta strains and Spt3, Spt7, Spt20/Ada5, Ada2, and Gcn5 all copurify with this nucleosomal HAT complex. Therefore, the 1.8-MD adaptor/HAT complex illustrates an interaction between Ada and Spt gene products and confirms the existence of a complex containing the TBP group of Spt proteins as demonstrated by genetic and biochemical studies. We have named this novel transcription regulatory complex SAGA (Spt-Ada-Gcn5-Acetyltransferase). The function of Gcn5 as a histone acetyltransferase within the Ada and SAGA adaptor complexes indicates the importance of histone acetylation during steps in transcription activation mediated by interactions with transcription activators and general transcription factors (i.e., TBP).
Article
In this paper we review the existing evidence that peritoneally derived B-1 cells may contribute significantly to the generation of IgA-secreting plasma cells in the murine intestinal lamina propria. The evidence is based upon a variety of experimental approaches performed in our laboratory and others and include (i) transfer studies of (sorted) B-1 cells into B-cell-depleted mice either experimentally (X-irradiation, anti-μ treatment) or genetically (SCID), (ii) analysis of genetically modified or manipulated mice (motheaten mice, CBA/N Xid mice, μ,k transgenic mice), and (iii) transplantation studies of fetal omentum. The data thus support the view that in addition to conventional B cells (B-2 cells) located in the Peyer′s patches (PP), B-1 cells contribute to the pool of IgA containing cells in the gut. Indeed, cotransfer of PP cells and peritoneal cells (PerC), which contain largely B-1 cells, into SCID recipients demonstrates that both PP and PerC contribute in a balanced fashion to the pool of IgA-containing cells in the gut lamina propria over long periods. Most likely IgA-positive (memory) B cells in PP are responsible for the long-term generation of IgA-producing cells derived from the PP inoculum. The potency of B-1 cells to contribute to mucosal IgA responses is also illustrated in adoptive transfer experiments in which PerC B-1 cells (or sorted B-1 cells) are adoptively transferred into untreated, Ig allotype congenic, SCID mice. These studies show that 6 months after injection of a few million PerC, almost all B cells in spleen and the recipient′s peritoneal cavity have the B-1 cell phenotype, while approximately 40 million PerC donor-derived IgA-producing cells can be detected in the gut lamina propria by allotype-specific ELISA spot assays. In conclusion, the data presented here show that, in principle, B-1 cells located in the peritoneal cavity may be an important source of precursors for intestinal IgA plasma cells in the mouse. However, the experiments performed so far do not allow us to draw definitive conclusions yet on the physiological contribution (in terms of numbers) and function (in terms of their specificity repertoire) of both B-1 cells and Peyer′s patch-derived B-2 cells to the pool of IgA-containing cells in the gut lamina propria in normal, unmanipulated animals.
Article
The c-Myc protein functions as a transcription factor to facilitate oncogenic transformation; however, the biochemical and genetic pathways leading to transformation remain undefined. We demonstrate here that the recently described c-Myc cofactor TRRAP recruits histone acetylase activity, which is catalyzed by the human GCN5 protein. Since c-Myc function is inhibited by recruitment of histone deacetylase activity through Mad family proteins, these opposing biochemical activities are likely to be responsible for the antagonistic biological effects of c-Myc and Mad on target genes and ultimately on cellular transformation.
Article
The phosphatidylinositol kinase-related (PIK-related) kinases form a new subgroup within the protein kinase family of enzymes. Although the substrates of these kinases are not yet known, recent reports show that they participate in recombination processes, chromosome maintenance, and the cell cycle.
Article
This chapter focuses on the transformation of primary rat embryo cells. Carcinogenesis is a multistep process involving the activation of oncogenes and the loss or inactivation of tumor suppressor genes. The transfection of certain combinations of distinct cellular oncogenes (for example, ras and myc) or cellular and viral oncogenes (for example, ras and adenovirus E1A) is required to induce tumorigenic conversion. The rat embryo cell cotransformation assay provides an invaluable tool to score the biological activity of these genes easily. The preparation of rat embryo cells and the cotransformation assay are described and materials and reagents required are also highlighted.
Article
The yeast transcriptional adaptor, Gcn5p, is a catalytic subunit of a nuclear (type A) histone acetyltransferase linking histone acetylation to gene activation. Here we report that Gcn5p acetylates histones H3 and H4 non-randomly at specific lysines in the amino-terminal domains. Lysine 14 of H3 and lysines 8 and 16 of H4 are highly preferred acetylation sites for Gcn5p. We also demonstrate that lysine 9 is the preferred position of acetylation in newly synthesized yeast H3 in vivo. This finding, along with the fact that lysines 5 and 12 in H4 are predominant acetylation sites during chromatin assembly of many organisms, indicates that Gcn5p acetylates a distinct set of lysines that do not overlap with those sites characteristically used by type B histone acetyltransferases for histone deposition and chromatin assembly.
Article
p300/CBP is a transcriptional adaptor that integrates signals from many sequence-specific activators via direct interactions. Various cellular and viral factors target p300/CBP to modulate transcription and/or cell cycle progression. One such factor, the cellular p300/CBP associated factor (PCAF), possesses intrinsic histone acetyltransferase activity. Here, we demonstrate that p300/CBP is not only a transcriptional adaptor but also a histone acetyltransferase. p300/CBP represents a novel class of acetyltransferases in that it does not have the conserved motif found among various other acetyltransferases. p300/CBP acetylates all four core histones in nucleosomes. These observations suggest that p300/CBP acetylates nucleosomes in concert with PCAF.