ArticlePDF Available

Evidence for a transcriptional activation function of BRCA1 C-terminal region

Authors:

Abstract and Figures

Mutations in BRCA1 account for 45% of families with high incidence of breast cancer and for 80-90% of families with both breast and ovarian cancer. BRCA1 protein includes an amino-terminal zinc finger motif as well as an excess of negatively charged amino acids near the C terminus. In addition, BRCA1 contains two nuclear localization signals and localizes to the nucleus of normal cells. While these features suggest a role in transcriptional regulation, no function has been assigned to BRCA1. Here, we show that the C-terminal region, comprising exons 16-24 (aa 1560-1863) of BRCA1 fused to GAL4 DNA binding domain can activate transcription both in yeast and mammalian cells. Furthermore, we define the region comprising exons 21-24 (aa 1760-1863) as the minimal transactivation domain. Any one of four germ-line mutations in the C-terminal region found in patients with breast or ovarian cancer (Ala-1708-->Glu, Gln-1756 C+, Met-1775-->Arg, Tyr-1853 ->Stop), had markedly impaired transcription activity. Together these data underscore the notion that one of the functions of BRCA1 may be the regulation of transcription.
Content may be subject to copyright.
Proc. Natl. Acad. Sci. USA
Vol. 93, pp. 13595–13599, November 1996
Biochemistry
Evidence for a transcriptional activation function of BRCA1
C-terminal region
(breast cancer
y
tumor suppressor gene)
ALVARO N. A. MONTEIRO*, AVERY AUGUST*, AND HIDESABURO HANAFUSA
Laboratory of Molecular Oncology, The Rockefeller University, New York, NY 10021
Contributed by Hidesaburo Hanafusa, September 11, 1996
ABSTRACT Mutations in BRCA1 account for 45% of
families with high incidence of breast cancer and for 80–90%
of families with both breast and ovarian cancer. BRCA1
protein includes an amino-terminal zinc finger motif as well
as an excess of negatively charged amino acids near the C
terminus. In addition, BRCA1 contains two nuclear localiza-
tion signals and localizes to the nucleus of normal cells. While
these features suggest a role in transcriptional regulation, no
function has been assigned to BRCA1. Here, we show that the
C-terminal region, comprising exons 16–24 (aa 1560–1863) of
BRCA1 fused to GAL4 DNA binding domain can activate
transcription both in yeast and mammalian cells. Further-
more, we define the region comprising exons 21–24 (aa
1760–1863) as the minimal transactivation domain. Any one
of four germ-line mutations in the C-terminal region found in
patients with breast or ovarian cancer (Ala-1708 3Glu,
Gln-1756 C1, Met-1775 3Arg, Tyr-1853 3Stop), had
markedly impaired transcription activity. Together these data
underscore the notion that one of the functions of BRCA1 may
be the regulation of transcription.
Breast cancer is one of the most common diseases affecting
women in the United States. Genetic factors contribute to an
estimated 5% of all breast cancer cases, and up to 36% of the
cases diagnosed before age 30 (1). In 1990, human BRCA1 was
mapped by genetic linkage to the long arm of chromosome 17
(2), firmly establishing the existence of a breast cancer sus-
ceptibility gene. Mutations in BRCA1 alone account for '45%
of the families with high incidence of breast cancer and 80%
of families with high incidence of both breast and ovarian
cancer (3). Identification of human BRCA1 by positional
cloning techniques revealed an ORF coding for 1863 aa (4). No
homology with any known protein was identified with the
exception of a zinc finger domain located in its N-terminal
region. This zinc binding RING finger domain (C3HC4) is
found in several proteins that have their functions mediated
through DNA binding (5). Other features in the sequence were
also recognized, including two putative nuclear localization
signals (aa 500–508 and 609– 615), a leucine zipper (aa 1209
1231), and an excess of negative charged residues in the
C-terminal region of BRCA1. In many eukaryotic transcrip-
tion activators, the presence of an acidic region in the C
terminus correlates with the transactivation domain (6). With
the exception of the leucine zipper, which is imperfectly
conserved, the other features are highly conserved in human
and mouse Brca1, suggesting that these regions might be
significant for its functions (7–9). Although reports have been
published showing that BRCA1 localizes to the cytoplasm and
secretory granules (10, 11), other investigators have shown that
BRCA1 localizes to the nucleus of normal cells (12, 13). Taken
together these features suggest a function of BRCA1 in
transcription activation.
We have investigated whether the C-terminal region of
BRCA1 is able to activate transcription. We found that
BRCA1 C terminal can act as a transactivation domain when
fused to a heterologous DNA binding domain, and that
germ-line mutations found in patients impair this activity,
suggesting that loss of transcription activation function by
BRCA1 may predispose the carriers to cancer.
MATERIALS AND METHODS
Yeast Strains. Two Saccharomyces cerevisiae strains, HF7c
[MATa, ura3-52, his3-200, lys2-801, ade2-101, trp1-901, leu2-3,
112, gal4-542, gal80-538, LYS::GAL1-HIS3, URA3::(GAL4 17-
mers)
3
-CYC1-lacZ] (14) and SFY526 (MATa, ura3-52, his3-
200, lys2-801, ade2-101, trp1-901, leu2-3, 112, can
r
, gal4-542,
gal80-538, URA3::GAL1-lacZ) (15), were used (CLON-
TECH). HF7c has a HIS3 reporter gene under the control of
the GAL1 upstream activating sequence (UAS), responsive to
GAL4 transcriptional activation. The HF7c transformants
were inoculated in liquid medium lacking either tryptophan or
tryptophan and histidine. The vectors used for expression
confer growth in the absence of tryptophan (see below). If the
fusion proteins activate transcription, yeast transformants are
able to grow in medium lacking histidine. HF7c also has a lacZ
reporter gene under the control of GAL4 17-mers UAS,
responsive to GAL4 transcriptional activation. The SFY526
strain has lacZ under the control of GAL1 UAS. If the fusion
proteins activate transcription, yeast transformants will pro-
duce
b
-galactosidase.
Yeast Expression Constructs. All BRCA1 fragments were
amplified by PCR using the plasmid F3 (a kind gift from
Robert Bogden, Myriad Genetics, Salt Lake City) as a tem-
plate, which contains the C-terminal domain of human BRCA1
(4). The following oligonucleotide primers were used: exons
16–24 (S9503101 and Z9503098, both containing an EcoRI
site 59and a BamHI site 39), exons 16–23 (S9503101 and L23),
exons 16–22 (S9503101 and LX22), exons 16–21 (S9503101
and Z9403097) and exons 22–24 (Z9503100 and Z9503098),
exons 21–24 (S9600330 and Z9503098), exons 21–22 (S9600330
and LX22) (S9503101, 59-CGGAATTCGAGGGAACCCCT-
TACCTG-39; Z9503098, 59-GCGGATCCGTAGTGGCTGT-
GGGGGAT-39; L23, 59-GCGGATCCTGCATGGAAGC-
CATTGTCC-39; LX22, 59-CGGGATCCACCTGTGC-
CAAGG-39; Z9403097, 59-GCGGATCCATCTGTGGGC-
ATGTTGGT-39; Z9503100, 59-CGGAATTCCAACTGGAAT-
GGATGGTA-39; S9600330, 59-CGGAATTCCAGGACAGA-
AAGATC-39). PCR products were gel purified, digested with
BamHI and EcoRI, and ligated into the yeast expression vector
pGBT9 (CLONTECH) (16) similarly digested. Fragments were
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
Abbreviations: X-Gal, 5-bromo-4-chloro-3-indolyl
b
-D-galctopyrano-
side; SD, synthetic medium plus dextrose.
*A.N.A.M. and A.A. contributed equally to this work.
To whom reprint requests should be addressed at: Laboratory of
Molecular Oncology, Box 169, The Rockefeller University, 1230
York Avenue, New York, NY 10021.
13595
ligated in frame to the GAL4 DNA binding domain. The junc-
tions were sequenced to verify the reading frame. pGBT9 has
TRP1 as a selectable marker, allowing growth in the absence of
tryptophan. For the TA 16y24, the construct 16y24 pGBT9 was
digested with BamHI and EcoRI, and the insert subcloned
in-frame to the GAL4 transactivation domain in pGAD424
(CLONTECH) (16) similarly digested. pGAD424 has LEU2 as a
selectable marker, allowing growth in the absence of leucine.
Mutations were made using splicing by overlap extension
PCR (17) with the following oligonucleotide primers: for the
Ala-1708 3Glu mutation S9503101 and LAE; UAE and
Z9503098 in two separate reactions. Both products were
combined and used as a template for a final round of PCR
using S9503101 and Z9503098. All subsequent mutations used
the same oligonucleotides (S9503101 and Z9503098) for the
final round of PCR. For the first round of PCR the following
oligonucleotides were used: for the Gln-1756 C1mutation
Z9600205 and S9503101; Z9600204 and Z9503098. For the
Met-1775 3Arg mutation S9503101 and S9600207; S9600206
and Z9503098. For the Met-1775 3Lys, Met-1775 3Thr,
Met-1775 3Glu, and Met-1775 3Val the S9600207 was
replaced by primers LMK, LMT, LME, and LMV, respec-
tively; and S9600206 was replaced by primers UMK, UMT,
UME, and UMV, respectively. The Tyr-1853 3Stop mutation
was obtained by direct PCR using S9503101 and L1853 (con-
taining a BamHI site) (LAE, 59-CCCATTTTCCTCCCT-
CAATTCCTAGAAAA-39; UAE, 59-TTTTCTAGGAATT-
GAGGGAGGAAAATGGG-39; Z9600205, 59-GATCTT-
TCTGTCGCTGGGATTCTCTTGCTC-39; Z9600204, 59-
GAGCAAGAGAATCCCAGCGACAGAAAGATC-39;
S9600207, 59-CAGTTGATCTGTGGGCCTGTTGGT-
GAAGG-39; S9600206, 59-CCTTCACCAACAGGCCCACA-
GATCAACTG-39; LMK, 59-CAGTTGATCTGTGGGCTT-
GTTGGTGAAGG-39; UMK, 59-CCTTCACCAACAAGC-
CCACAGATCAACTG-39; LMT, 59-CAGTTGATCTGTG-
GGCGTGTTGGTGAAGG-39; UMT, 59-CCTTCACCAAC-
ACGCCCACAGATCAACTG-39; LME, 59-CAGTTGATCT-
GTGGGCTCGTTGGTGAAGG-39; UME, 59-CCTTCAC-
CAACGAGCCCACAGATCAACTG-39; LMV, 59-CAGTT-
GATCTGTGGGCACGTTGGTGAAGG-39; UMV, 59-
CCTTCACCAACGTGCCCACAGATCAACTG-39; L1853,
59-GCGGATCCAGGTTAGGTGTCC-39). The constructs
were sequenced to confirm the mutations.
Yeast Transformation. Competent yeast cells were obtained
using the yeast transformation system (CLONTECH) based on
lithium acetate, and cells were transformed according to
manufacturer’s instructions.
Interference Assay. Levels of expression were assessed by
transforming the HF7c strain with pCL1 (wild-type full-length
GAL4) (18) and measuring reduction in transcription activa-
tion by pCL1 upon cotransformation with the pGBT9-based
constructs. Three independent clones from each construct
were assayed for
b
-galactosidase production in liquid culture
as described (19).
Growth Curves. HF7c transformants containing different
pGBT9 constructs were grown overnight in synthetic medium
plus dextrose (SD medium) lacking tryptophan. The saturated
cultures were used to inoculate fresh medium lacking trypto-
phan or tryptophan and histidine to an initial OD
600
of 0.0002.
Cultures were grown at 308C in a shaker and the optical density
was measured at different time intervals starting at 12 hr, then
every 4 hr up to 36 hr after inoculation.
Growth on Solid Medium. HF7c transformants were
streaked on solid SDyagar medium lacking tryptophan or
tryptophan and histidine. Growth was scored after 2 days.
Filter
b
-Galactosidase Assay. SFY526 and HF7c transfor-
mants were streaked on a filter overlaid on top of medium
lacking tryptophan and allowed to grow overnight. Cells
growing on the filter were lysed by freeze thawing in liquid
nitrogen, and each filter was incubated in 2.5 ml of Z buffer
(16.1 g/liter Na
2
HPO
4
z7H
2
Oy5.5 g/liter NaH
2
PO
4
zH
2
Oy0.75
g/liter KCly0.246 g/liter MgSO
4
z7H
2
O, pH 7.0) containing 40
m
l of 5-bromo-4-chloro-3-indolyl
b
-D-galctopyranoside (X-
Gal) solution (20 mgyml of X-Gal in N,N-dimethylformamide)
at 308C for up to 24 hr.
Mammalian Expression Constructs: pGBT9-based con-
structs containing the wild-type 16y24 sequences as well as the
mutants were digested with EcoRI and XbaI, and the frag-
ments including vector sequences downstream of the insert
were ligated into similarly digested pSG424 (20) (a gift of Mark
Ptashne, Harvard University). The fragments were subcloned
in frame to GAL4 binding domain similar to the yeast expres-
sion vectors.
Transient Transfections. Cos-7 cells were grown until con-
fluent in DMEM (GIBCOyBRL) supplemented with 10%
fetal bovine serum. Cells were transfected using Lipo-
fectamine (GIBCOyBRL). Briefly, confluent cells were
washed with OptiMEM (GIBCOyBRL) and overlaid with a
mixture containing the following plasmids and Lipofectamine:
0.4
m
g of pSG424 or pSG424 containing 16y24 wild-type,
16y24 (Gln-1756 C1) and 16y24 (Met-1775 3Arg), all
together with 1
m
g of reporter pG5E1bLuc (a gift of Roger
Davis, University of Massachusetts) (21). Cells were exposed
to the transfection mixture for 4 hr, then washed and subjected
to a 2-min 15% glycerol shock. After the shock, fresh medium
was added and cells were harvested 24 and 48 hr after
transfection. Cells were lysed and extracts were assayed for
luciferase activity as described (21).
RESULTS
Transcription Activation by BRCA1 C-Terminal Region.
The vast majority of reported mutations in BRCA1 lead
invariably to truncation of the C-terminal region. The few
missense mutations reported localize either to the zinc finger
motif at the N terminus or to the C-terminal acidic region (22).
Moreover, the C-terminal region includes a highly conserved
stretch between mouse and human BRCA1 (aa 1636 –1795) (8).
We therefore investigated whether the region of a reported
cluster of mutations adjacent to the C-terminal end (22),
comprising exons 16–24 (aa 1560–1863), is involved in tran-
scriptional activation.
Because BRCA1 DNA binding consensus has not been
defined and increasing evidence suggests that many cellular
processes, including transcription, are conserved among eu-
karyotic species, a yeast GAL4 DNA binding domain fusion
system was used (16). We fused the GAL4 DNA binding
domain with human BRCA1 fragments containing either
exons 16y24, 16y23, 16y22, 16y21, 22y24, 21y24, or 21y22. The
16y24 fragment fused to transactivation domain of GAL4,
which is unable to bind DNA (TA-16y24) was used as a
negative control (Fig. 1). These constructs were transformed
into yeast and ability to activate the reporter genes was tested.
As shown in Fig. 2, only the fusions 16y24 and 21y24 were able
to activate transcription of the HIS3 reporter gene. The 21y24
construct however, activated transcription somewhat less ef-
ficiently than the larger 16y24. Scoring HF7c transformants for
growth on solid media gave identical results (Table 1). In
agreement with the results obtained w ith HF7c, only 16y24 and
21y24 were able to activate transcription of lacZ in SFY526
(Table 1). No construct was able to activate
b
-galactosidase
production in HF7c cells (data not shown). The 16y24 con-
struct is thus able to function as a transactivation domain and
sequences contained within exons 21 to 24 of BRCA1 have
sufficient information for activation of transcription, although
sequences contained in adjacent exons may enhance this
activity.
Germ-Line Mutations Impair Transcription Activation. It
has been shown that retroviral transfer of wild-type BRCA1
gene inhibits growth in vitro of breast and ovarian cancer cell
13596 Biochemistry: Monteiro et al. Proc. Natl. Acad. Sci. USA 93 (1996)
lines, thus establishing that BRCA1 acts as a tumor suppressor
(23). Loss or reduction of function of BRCA1 would therefore
confer susceptibility to tumor progression. The fact that a
portion of BRCA1 protein was able to drive expression of the
reporter genes raised the possibility that impairment of this
function could be related to the development of breast and
ovarian cancer.
To test this hypothesis directly we introduced four mutations
reported in afflicted patients, generating the constructs 16y24
(Ala-1708 3Glu, Gln-1756 C1, Met-1775 3Arg, and
Tyr-1853 3Stop) shown in Fig. 1. Ala-1708 3Glu is
associated with very early onset of breast cancer (24). Gln-1756
C1is an insertion of a cytosine in codon 1756 resulting in a
frameshift and is the most frequent mutation found in the
C-terminal (4, 25–29). Met-1775 3Arg is the only recurring
missense mutation in this region (4, 22, 24, 26). Moreover, this
residue lies in a highly conserved stretch between mouse and
human BRCA1 (8). Tyr-1853 3Stop is an insertion of an
adenine in codon 1853 leading to a truncated protein only 11
aa from the C-terminal end that is associated with very
early-onset breast cancer (30). None of the constructs carrying
mutations found in patients was able to activate transcription
of the HIS3 promoter in HF7c (Fig. 3), nor lacZ expression in
SFY526 (Table 1). The difference in activation between the
wild type and the mutants cannot be accounted for different
levels in expression and stability as all constructs were ex-
pressed at roughly the same levels as measured by interference
assays (data not shown).
Transcription Activation in Mammalian Cells. To rule out
the possibility that BRCA1 activates transcription only in the
yeast system, we examined the transcriptional activity of the
wild-type 16y24 and two mutants (Gln-1756 C1and Met-1775
3Arg) using a mammalian cell line, Cos-7. In this system we
used GAL4 DNA binding domain fusions (20) and the lucif-
erase gene as a reporter under the control of GAL4 responsive
elements (21). In Fig. 4 the wild-type fusion activates tran-
scription 10-fold, while the mutants were not able to activate
transcription to a detectable level. Therefore, BRCA1 tran-
scriptional activation function is not restricted to the yeast
system. Similarly to the experiments performed in yeast, the
difference cannot be accounted for different levels of expres-
sion, since all constructs were expressed, with the Met-1775 3
Arg expressed at higher levels than the others, as determined
by Western blotting (data not shown).
FIG. 1. Structure of fusion proteins. Fragments 16y24, 16y23,
16y22, 16y21, 21y24, 22y24, and 21y22 contain exons 16 –24 (aa
1560–1863, top bar), 16 –23, 16–22, 16–21, 21–24, 22–24, and 21–22,
respectively, of the BRCA1 gene fused to the DNA binding domain of
GAL4 (black box). Fragments 16y24 (Gln-1756 C1) and 16y24
(Tyr-1853 3Stop) carry insertions reported to truncate BRCA1.
Fragment 16y24 (Ala-1708 3Glu) is a substitution reported in
patients. Fragments 16y24 (Met-1775 3Lys, Met-1775 3Thr,
Met-1775 3Arg, Met-1775 3Glu, and Met-1775 3Val) are each
individual mutations at codon 1775, with the Met-1775 3Arg mutant
reported in patients with breast cancer. TA 16y24 represents exons
16–24 fused to the activation domain of GAL4. Arrowheads indicate
the positions of the mutations, and shaded areas represent truncated
parts of the mutant protein.
FIG. 2. Transcriptional activation by BRCA1 C terminus. Growth
curves of S. cerevisiae (HF7c) carrying the following fragments of
BRCA1 in pGBT9 grown in medium lacking tryptophan and histidine:
16y24 (
M
), 16y23 (
m
), 16y22 (
E
), 16y21 (
), 21y24 (
Ç
), 22y24 (
å
), and
21y24 (
). (Inset) Growth in medium lacking tryptophan only. OD
600
of the cultures (yaxis) is plotted against time in culture (xaxis).
Table 1. Transcriptional activation by BRCA1 C terminus
HF7c SFY526
b
-galCOS-7
luciferaseLiquid* Solid
Wild type
16y24 1(1.0) 111(1.0)
16y23 2(0.0) 22 ND
16y22 2(0.0) 22 ND
16y21 2(0.0) 22 ND
21y24 1(0.6) 11 ND
22y24 2(0.0) 22 ND
21y22 2(0.0) 22 ND
16y24-TA ND 22 ND
16y24 mutants
Ala-1708 3Glu 2(0.0) 22 ND
Gln-1756 C12(0.0) 222(0.0)
Met-1775 3Arg 2(0.0) 222(0.0)
Tyr-1853 3Stop 2(0.0) 22 ND
Met-1775 3Lys 2(0.0) 22 ND
Met-1775 3Val 1(1.0) 11 ND
Met-1775 3Thr 2(0.0) 21 ND
Met-1775 3Glu 2(0.0) 22 ND
ND, not determined.
*Cultures were grown as described in Materials and Methods. Relative
activity shown in parenthesis.
Several independent colonies were streaked on SD agar lacking
tryptophan and histidine and scored for growth after 3 days at 308C.
Several independent clones were assayed for
b
-galactosidase (
b
-gal)
production on filters. To ensure that the negatives were not producing
b
-galactosidase, the assay was also scored after 18 h with the same
results.
Biochemistry: Monteiro et al. Proc. Natl. Acad. Sci. USA 93 (1996) 13597
Hydrophobic Residues Are Major Determinants. Acidic
regions of proteins that are not necessarily transcription acti-
vators can sometimes activate transcription when fused to
heterologous DNA binding domains (31). To exclude this
possibility in our assay using BRCA1 C terminus, we intro-
duced a negative charge at position 1775 (Met-1775 3Glu).
As shown in Table 1 this construct was not able to activate
transcription, eliminating the possibility that a nonspecific
cluster of negative charges in BRCA1 was spuriously activating
transcription.
As has been suggested in other acidic domain-containing
transcription factors, addition of positively charged residues
irrespective of the position results in impairment of the
transcriptional activation function (32). The mutant Met-1775
3Arg described above is a change from a hydrophobic to that
of a positively charged residue, which resulted in drastically
impaired activity of BRCA1 C terminus. We examined the
effects of three other possible mutations at codon 1775: from
the hydrophobic methionine to (i) a positively charged residue
change similar to that seen in the reported patients (Met-1775
3Lys), (ii) a polar residue (Met-1775 3Thr), and (iii)a
hydrophobic residue (Met-1775 3Val), for both growth in
liquid and solid media, as well as in
b
-galactosidase assays.
Changing the methionine at position 1775 to a conservative
residue valine, resulted in wild-type transcriptional activity,
while a change to a positively charged residue (Met-1775 3
Lys) or to a polar residue (Met-1775 3Thr) drastically
diminished transcriptional activation by BRCA1 C terminus
(Table 1). Interestingly, the construct carrying the Met-1775 3
Thr mutation does not change the net charge of the molecule
and is still inactive in HF7c. These data suggest that BRCA1
may function analogously to herpes virus protein VP16 where
negative charges are not the only major determinants but that
hydrophobic residues are also crucial for activity (33).
DISCUSSION
The presence of an acidic region, nuclear localization signals
and a zinc finger domain suggest that BRCA1 may regulate
transcription (4). By using fusion proteins with a heterologous
DNA binding domain (GAL4), we show that the C-terminal
portion of BRCA1 (aa 1560–1863) is able to activate tran-
scription of the HIS3 gene in HF7c (but not lacZ), the lacZ
gene in SFY526, as well as the luciferase gene in mammalian
cells, all driven by GAL4 responsive elements. Although HIS3
and lacZ in HF7c share GAL4-responsive elements, the rest of
the promoter sequences are different (14, 15). The lack of
transcriptional activation of lacZ in HF7c may therefore
reflect the fact that the ability of transcriptional activators to
interact with the basal transcription apparatus varies with
different promoter contexts (34, 35).
Our results also indicate that the smaller C-terminal frag-
ment 21y24 (aa 1760–1863) is also able to activate transcrip-
tion, although less efficiently than the larger fragment 16y24
(aa 1560–1863). Therefore, exons 21y24 have sufficient infor-
mation to activate transcription and represent the minimal
transactivation domain of BRCA1. Moreover, adjacent exons,
16 to 20, contribute to full activity. Interestingly, most muta-
tions found in patients generate proteins lacking all or part of
the minimal transactivation domain defined above. According
to our data, truncated proteins lacking even small regions of
the minimal transactivation domain (e.g., Tyr-1853 3Stop)
would be predicted to have no function in transcriptional
activation, a situation borne out by our data.
While the wild-type BRCA1 C terminus fused to a heter-
ologous DNA binding protein activates transcription, the
proteins carrying missense or nonsense mutations found in
patients with tumors fail to do so. The fact that fusion proteins
carrying mutations known to confer breast cancer susceptibil-
ity abolish activity strongly supports the idea that BRCA1
protein is involved in transcription. We tested four reported
mutations (Ala-1708 3Glu, Gln-1756 C1, Met-1775 3Arg,
and Tyr-1853 3Stop) with association with disease (4, 22,
24–29). Thus we suggest that mutations in BRCA1 which
impair the ability to activate transcription may predispose the
carrier to tumors. These results indicate that BRCA1 may
function to activate transcription of genes involved in sup-
pressing transformation.
Interestingly, a mutation that falls outside the minimal
transactivation domain also abolished function (Ala-1708 3
Glu). The observation that a construct lacking this region is
active suggests that it may be involved in proper folding of the
C-terminal region, which may either stabilize or destabilize the
structure. When the region is deleted, the conformation may
be conserved as in the wild type. An alternative explanation is
that this amino acid is involved in nonessential sites of protein–
protein interactions and that absence of these sites or nondis-
ruptive mutations allow interaction. Other mutations such as
FIG. 3. Mutations occurring in patients impair activity. Shown are
growth curves of S. cerevisiae (HF7c) carrying the following fragments
and mutants in pGBT9 grown in medium lacking tryptophan and
histidine: 16y24 (
M
), 16y24 (Ala-1708 3Glu) (
å
), 16y24 (Gln-1756
C1)(
E
), 16y24 (Met-1775 3Arg) (
), and 16y24 (Tyr-1853 3Stop)
(
). (Inset) Growth in medium lacking tryptophan only. The OD
600
of
the cultures (yaxis) is plotted against time in culture (xaxis).
FIG. 4. Transcription activation by BRCA1 C terminus in mam-
malian cells. Luciferase activity of the following fragments and mu-
tants in pSG424, transfected along with luciferase reporter plasmid
into Cos-7 cells plotted as fold activation over control: vector, pSG424
alone; 16y24, wild-type exons 16y24; 16y24 (Met-1775 3Arg),
Met-1775 3Arg mutation; 16y24 (Gln-1756 C1), cytosine insertion
at position 1756. Cells were harvested at 24 hr (black bars) and 48 hr
(shaded bars).
13598 Biochemistry: Monteiro et al. Proc. Natl. Acad. Sci. USA 93 (1996)
Ala-1708 3Glu on the other hand, may destabilize this
interaction.
During the preparation of this manuscript, Chapman and
Verma (36) reported similar results. Using a mammalian
transfection system the authors defined the fragment 16y24 as
a transactivation domain and suggested that the fragment
21y24 represents the minimal transactivation domain, in
agreement with our data. Taken together, these two studies
analyze five different mutations of BRCA1, and all the mu-
tations which have shown disease association have impaired
ability to activate transcription. We believe that a detailed
analysis of BRCA1 C-terminal region may be instrumental to
understanding the etiology of familial breast cancer.
The simple assay used here, testing the ability of the BRCA1
fusion proteins to activate transcription in yeast is a direct way
to assess the functional significance of documented mutations
that localize to the transactivation domain. Furthermore, our
system could be instrumental in discriminating between cancer
predisposing mutations and neutral polymorphisms. The assay
could also be used to screen for therapeutic compounds
capable of restoring the normal function to the mutant pro-
teins.
Definite confirmation that BRCA1 acts as a transcription
activator, however, must await the demonstration of its DNA
binding activity or identification of its target genes.
We thank Alison Egan for help with the constructs. We also thank
Robert Roeder, Alexander Hoffmann, and Curt Horvath for helpful
discussions and Shinya Tanaka and Raymond Birge for critical com-
ments on the manuscript. A.N.A.M. is a Pew Fellow in the Biomedical
Sciences and on leave from The Institute of Chemistry, Federal
University of Rio de Janeiro. A.A. is a recipient of a post-doctoral
fellowship from the National Science Foundation. This work was
supported by Grant CA44356 from the National Cancer Institute.
1. Claus, E. B., Risch, N. & Thompson, W. D. (1991) Am. J. Hum.
Genet. 48, 232–242.
2. Hall, J. F., Lee, M. K., Newman, B., Morrow, J. E., Anderson,
L. A., Huey, B. & King, M.-C. (1990) Science 250, 1684–1689.
3. Easton, D. F., Bishop, D. T., Ford, D., Crockford, G. P. & Breast
Cancer Linkage Consortium (1993) Am. J. Hum. Genet. 52,
678–701.
4. Miki, Y., Swensen, J., Shattuck-Eidens, D., Futreal, P. A., Harsh-
man, K., et al. (1994) Science 266, 66–71.
5. Saurin, A. J., Borden, K. L. B., Boddy, M. N. & Freemont, P. S.
(1996) Trends Biochem. Sci. 21, 208–214.
6. Ptashne, M. (1988) Nature (London) 335, 683–689.
7. Lane, T. F., Deng, C., Elson, A., Lyu, M. S., Kozak, C. A. &
Leder, P. (1995) Genes Dev. 9, 2712–2722.
8. Abel, K. J., Xu, J., Yin, G.-Y., Lyons, R. H., Meisler, M. H. &
Weber, B. L. (1995) Hum. Mol. Genet. 4, 2265–2273.
9. Sharan, S. K., Wims, M. & Bradley, A. (1995) Hum. Mol. Genet.
4, 2275–2278.
10. Jensen, R. A., Thompson, M. E., Jetton, T. L., Szabo, C. I., van
der Meer, R., Helou, B., Tronick, S. R., Page, D. L., King, M.-C.
& Holt, J. T. (1996) Nat. Genet. 12, 303–308.
11. Jensen, R. A., Thompson, M. E., Jetton, T. L., van der Meer, R.,
Helou, B., Arteaga, C. L., Page, D. L., Holt, J. T., Tronick, S. R.,
Gown, A. M., Skelly, M., Ostermeyer, B., Schieltz, D., Szabo,
C. I. & King, M.-C. (1996) Nat. Genet. 13, 269–272.
12. Chen, Y., Chen, C.-F., Riley, D. J., Allred, D. C., Chen, P.-L., von
Hoff, D., Osborne, C. K. & Lee, W.-H. (1995) Science 270,
789–791.
13. Scully, R., Ganesan, S., Brown, M., De Caprio, J. A., Cannistra,
S. A., Feunteun, J., Schnitt, S. & Liv ingston, D. M. (1996) Science
272, 123–125.
14. Bartel, P. L., Chien, C.-T., Sternglanz, R. & Fields, S. (1993)
BioTechniques 14, 920–924.
15. Feilotter, H. E., Hannon, G. J., Ruddel, C. J. & Beach, D. (1994)
Nucleic Acids Res. 22, 1502–1503.
16. Bartel, P. L., Chien, C.-T., Sternglanz, R. & Fields, S. (1993) in
Cellular Interactions in Development: A Practical Approach, ed.
Hartley, D. A. (Oxford Univ. Press, Oxford), pp. 153–179.
17. Higuchi, R. (1990) in PCR Protocols: A Guide to Methods and
Applications, eds. Innis, M. A., Gelfand, D. H., Sininski, J. J. &
White, T. J. (Academic, San Diego), pp. 177–183.
18. Fields, S. & Song, O. (1989) Nature (London) 340, 245–247.
19. Brent, R. & Ptashne, M. (1984) Nature (London) 312, 612–615.
20. Sadowski, I. & Ptashne, M. (1989) Nucleic Acids Res. 17, 7539.
21. Seth, A., Gonzalez, F. A., Gupta, S., Raden, D. L. & Davis, R. J.
(1992) J. Biol. Chem. 267, 24796–24804.
22. Breast Cancer Information Core, http:yywww.nchgr.nih.govy
dirylab_transferybicy.
23. Holt, J. T., Thompson, M. E., Szabo, C., Robinson-Benion, C.,
Arteaga, C. L., King, M.-C. & Jensen, R. A. (1996) Nat. Genet.
12, 298–302.
24. Futreal, P. A., Liu, Q., Shattuck-Eidens, D., Cochran, C., Harsh-
man, K., et al. (1994) Science 266, 120–122.
25. Simard, J., Tonin, P., Durocher, F., Morgan, K., Rommens, J.,
Gingras, S., Samson, C., Leblanc, J.-F., Belanger, C., Dion, F.,
Liu, Q., Skolnick, M., Goldgar, D., Shattuck-Eidens, D., Labrie,
F. & Narod, S. A. (1994) Nat. Genet. 8, 392–398.
26. Szabo, C. I. & K ing, M.-C. (1995) Hum. Mol. Genet. 4, 1811–1817.
27. Langston, A. A., Malone, K. E., Thompson, J. D., Daling, J. R. &
Ostrander, E. A. (1996) N. Engl. J. Med. 334, 137–142.
28. Gayther, S. A., Warren, W., Mazoyer, S., Russel, P. A., Har-
rington, P. A., Chiano, M., Seal, S., Hamoudi, R., van Rensburg,
E. J., Dunning, A. M., Love, R., Evans, G., Easton, D., Clayton,
D., Stratton, M. R. & Ponder, B. A. J. (1995) Nat. Genet. 11,
428– 433.
29. Serova, O., Montagna, M., Torchard, D., Narod, S. A., Tonin, P.,
Sylla, B., Lynch, H. T., Feunteun, J. & Lenoir, G. M. (1996)
Am. J. Hum. Genet. 58, 42–51.
30. Friedman, L. S., Ostermeyer, E. A., Szabo, C., Dowd, P. Lynch,
E. D., Rowell, S. E. & King, M.-C. (1994) Nat . Genet. 8, 399–403.
31. Ma, J. & Ptashne, M. (1987) Cell 51, 113–119.
32. Bushman, F. D. & Ptashne, M. (1988) Cell 54, 191–197.
33. Cress, W. D. & Triezenberg, S. J. (1991) Science 251, 87–90.
34. Estojak, J., Brent, R. & Golemis, E. A. (1995) Mol. Cell. Biol. 15,
5820–5829.
35. Seneca, S., Punyammalee, B., Bailly, M., Ghysdael, J. & Crabeel,
M. (1991) Oncogene 6, 357–360.
36. Chapman, M. S. & Verma, I. M. (1996) Nature (London) 382,
678– 679.
Biochemistry: Monteiro et al. Proc. Natl. Acad. Sci. USA 93 (1996) 13599
... Impaired assembly between the BRCT domain and interacting proteins has been shown to be associated with tumour susceptibility [16]. Accordingly, alterations in the BRCT domain can impair both the HRR and the TA activities of BRCA1 [17,18]. ...
... In this study, we have analysed the effect of 11 rare missense BRCA1 VUSs located within or in close proximity to the BRCT domain of BRCA1 with respect to protein expression, HRR and TA activity, with the aim of generating additional knowledge to guide the correct classification of these variants. The two C-terminal BRCT repeats of the BRCT domain of BRCA1 contain conserved hydrophobic/acidic patches, which have been shown to be crucial for both the HRR capacity and TA activity [17,18]. In addition, several missense variants located in the BRCT domain have previously been shown to destabilise folding of the BRCA1 protein, leading to increased premature protein degradation [26][27][28]. ...
Article
Full-text available
Background Damaging alterations in the BRCA1 gene have been extensively described as one of the main causes of hereditary breast and ovarian cancer (HBOC). BRCA1 alterations can lead to impaired homologous recombination repair (HRR) of double-stranded DNA breaks, a process which involves the RING, BRCT and coiled-coil domains of the BRCA1 protein. In addition, the BRCA1 protein is involved in transcriptional activation (TA) of several genes through its C-terminal BRCT domain. Methods In this study, we have investigated the effect on HRR and TA of 11 rare BRCA1 missense variants classified as variants of uncertain clinical significance (VUS), located within or in close proximity to the BRCT domain, with the aim of generating additional knowledge to guide the correct classification of these variants. The variants were selected from our previous study “BRCA1 Norway”, which is a collection of all BRCA1 variants detected at the four medical genetic departments in Norway. Results All variants, except one, showed a significantly reduced HRR activity compared to the wild type (WT) protein. Two of the variants (p.Ala1708Val and p.Trp1718Ser) also exhibited low TA activity similar to the pathogenic controls. The variant p.Trp1718Ser could be reclassified to likely pathogenic. However, for ten of the variants, the total strength of pathogenic evidence was not sufficient for reclassification according to the CanVIG-UK BRCA1/BRCA2 gene-specific guidelines for variant interpretation. Conclusions When including the newly achieved functional evidence with other available information, one VUS was reclassified to likely pathogenic. Eight of the investigated variants affected only one of the assessed activities of BRCA1, highlighting the importance of comparing results obtained from several functional assays to better understand the consequences of BRCA1 variants on protein function. This is especially important for multifunctional proteins such as BRCA1.
... Introduction of known disease-causing mutations within the BRCT domain, but not benign polymorph isms, has been shown to abolish this activity [113][114][115]. Yeast and! or mammalian assays of transcription activation have now been used to assess approximately 30 different missense mutations [115][116][117]. ...
... Such stUdies have confirmed that missense mutations with a deleterious effect on transcription activation are scattered throughout the BRCT coding region rather than being clustered in a particular "hot-spot" [115]. Importantly, one report noted discrepancies between results from yeast and mammalian based assays, suggesting that the simpler yeast-based assay may lack the complex transcription regulatory controls necessary to fully examine the effect of a human missense mutation on transcription [113]. [121], and has been limited to rely on only on immunohistochemical and pathological data, LOH and familial phenotype [122]. ...
Thesis
p>Over 450 distinct BRCA1 missense mutations have been found in patients with a family history of breast cancer and the functional significance of most of these is unclear. Increasing evidence suggests that DNA missense mutations can affect RNA stability or sequence by disrupting splicing regulators. I have used a variety of technique to investigate the effect of genomic BRCA1 missense mutations on transcript expression. BRCA1 monoallelic expression has previously been reported in association with missense mutations. I therefore initially used the technique of Pyrosequencing<sup>TM</sup> to identify imbalances in BRCA1 cDNA allele expression. Analysis of patients with known BRCA1 truncating mutations, missense mutations and controls identified no examples of monoallelic expression and indicated that a previous report of monoallelic BRCA1 expression was erroneous. I subsequently developed a series of multiplex RT-PCR reactions using overlapping primer pairs to identify alternative BRCA1 transcripts in the same groups of subjects. This technique effectively demonstrated the common BRCA1 isoforms and confirmed that the c4185A>G silent mutation is associated with deletion of exon 12. To provide a systematic analysis of the effect of BRCA1 mutations on splicing regulation I designed minigenes centred on 4 different BRCA1 exons: 5, 6, 10 and 18. All mutations within these exons reported to the Breast Cancer Information Core database were introduced into the appropriate minigenes, and wildtype and mutant minigenes were transfected into HEK 293 cells. The resulting transcripts were examined to identify aberrant splicing. Only one of twenty-one missense mutations investigated resulted in alternative transcripts, suggesting that only a small proportion of BRCA1 missense mutations do affect splicing. Additional work was also performed to investigate whether the MDM2 SNP 309 acts as a disease modifying gene in BRCA1 associated hereditary cancer.</p
... The black stars indicate significance of reduction compared to wt, and the red stars indicate differences between Ex20dup and individual variants with *p < 0.05, **p < 0.01 and, ***p < 0.001, ns: not significant suggested to be pathogenic with no further evidence provided for class assignment. However, exon 20 contains part of the BRCT domains, which mediate complex functions of BRCA1 in DNA damage response via, e.g., phospho-protein interactions [18] and in transcriptional activation [19,20]. Several known pathogenic missense variants have been reported in this domain including c.5213G > A p.(Gly1738Glu) in exon 20, illustrating the importance of this region [21]. ...
Article
Full-text available
Background Reports of dual carriers of pathogenic BRCA1 variants in trans are extremely rare, and so far, most individuals have been associated with a Fanconi Anemia-like phenotype. Methods We identified two families with a BRCA1 in-frame exon 20 duplication (Ex20dup). In one male individual, the variant was in trans with the BRCA1 frameshift variant c.2475delC p.(Asp825Glufs*21). We performed splicing analysis and used a transcription activation domain (TAD) assay to assess the functional impact of Ex20dup. We collected pedigrees and mapped the breakpoints of the duplication by long- and short-read genome sequencing. In addition, we performed a mitomycin C (MMC) assay from the dual carrier using cultured lymphoblastoid cells. Results Genome sequencing and RNA analysis revealed the BRCA1 exon 20 duplication to be in tandem. The duplication was expressed without skipping any one of the two exon 20 copies, resulting in a lack of wild-type transcripts from this allele. TAD assay indicated that the Ex20dup variant has a functional level similar to the well-known moderate penetrant pathogenic BRCA1 variant c.5096G > A p.(Arg1699Gln). MMC assay of the dual carrier indicated a slightly impaired chromosomal repair ability. Conclusions This is the first reported case where two BRCA1 variants with demonstrated functional impact are identified in trans in a male patient with an apparently normal clinical phenotype and no BRCA1-associated cancer. The results pinpoint a minimum necessary BRCA1 protein activity to avoid a Fanconi Anemia-like phenotype in compound heterozygous status and yet still predispose carriers to hormone-related cancers. These findings urge caution when counseling families regarding potential Fanconi Anemia risk. Furthermore, prudence should be taken when classifying individual variants as benign based on co-occurrence in trans with well-established pathogenic variants.
... After invading farmland, it competes with crops for water, light, fertilizer, and growth space. As a result, the yields of a variety of grains, oil crops, and cash crops are reduced to varying degrees, especially rice [36]. Due to the small row spacing of rice seedlings in paddy fields, weeds and seedlings can easily cover each other when growing in a limited space. ...
Article
Full-text available
Weeds are one of the greatest threats to the growth of rice, and the loss of crops is greater in the early stage of rice growth. Traditional large-area spraying cannot selectively spray weeds and can easily cause herbicide waste and environmental pollution. To realize the transformation from large-area spraying to precision spraying in rice fields, it is necessary to quickly and efficiently detect the distribution of weeds. Benefiting from the rapid development of vision technology and deep learning, this study applies a computer vision method based on deep-learning-driven rice field weed target detection. To address the need to identify small dense targets at the rice seedling stage in paddy fields, this study propose a method for weed target detection based on YOLOX, which is composed of a CSPDarknet backbone network, a feature pyramid network (FPN) enhanced feature extraction network and a YOLO Head detector. The CSPDarknet backbone network extracts feature layers with dimensions of 80 pixels ⊆ 80 pixels, 40 pixels ⊆ 40 pixels and 20 pixels ⊆ 20 pixels. The FPN fuses the features from these three scales, and YOLO Head realizes the regression of the object classification and prediction boxes. In performance comparisons of different models, including YOLOv3, YOLOv4-tiny, YOLOv5-s, SSD and several models of the YOLOX series, namely, YOLOX-s, YOLOX-m, YOLOX-nano, and YOLOX-tiny, the results show that the YOLOX-tiny model performs best. The mAP, F1, and recall values from the YOLOX-tiny model are 0.980, 0.95, and 0.983, respectively. Meanwhile, the intermediate variable memory generated during the model calculation of YOLOX-tiny is only 259.62 MB, making it suitable for deployment in intelligent agricultural devices. However, although the YOLOX-tiny model is the best on the dataset in this paper, this is not true in general. The experimental results suggest that the method proposed in this paper can improve the model performance for the small target detection of sheltered weeds and dense weeds at the rice seedling stage in paddy fields. A weed target detection model suitable for embedded computing platforms is obtained by comparing different single-stage target detection models, thereby laying a foundation for the realization of unmanned targeted herbicide spraying performed by agricultural robots.
... Several lines of evidence support the role for BRCA1 in transcriptional regulation. First, the BRCA1 C-terminal can activate transcription when fused to a heterologous DNA binding domain [42][43][44] . Second, BRCA1 is found in complex with the RNA Polymerase II holoenzyme and can bind DNA through specific sequences [45][46][47] . ...
Article
Full-text available
BRCA1 (Breast Cancer 1, early onset) is linked to breast and ovarian cancer predisposition. Still, the risks conferred by a significant portion of BRCA1 variants identified in the population remains unknown. Most of these variants of uncertain significance are missense alterations. However, the functional implications of small in-frame deletions and/or insertions (indels) are also difficult to predict. Our group has previously evaluated the functional impact of 347 missense variants using an extensively validated transcriptional activity assay. Here we show a systematic assessment of 30 naturally occurring in-frame indels located at the C-terminal region of BRCA1. We identified positions sensitive and tolerant to alterations, expanding the knowledge of structural determinants of BRCA1 function. We further designed and assessed the impact of four single codon deletions in the tBRCT linker region and six nonsense variants at the C-terminus end of BRCA1. Amino acid substitutions, deletions or insertions in the disordered region do not significantly impact activity and are not likely to constitute pathogenic alleles. On the other hand, a sizeable fraction of in-frame indels at the BRCT domain significantly impact function. We then use a Bayesian integrative statistical model to derive the probability of pathogenicity for each variant. Our data highlights the importance of assessing the impact of small in-frame indels in BRCA1 to improve risk assessment and clinical decisions for carriers.
Article
Transcription factors have proven to be promising targets for the treatment of cancer. Transcription factors are involved in the production of oxygen. External cervical events are initiated by receptors, such as cytotoxic exposures or cytokine receptors that trigger signalling cascades that activate transcription factors. Transcriptional factors are known to be highly active in most human cancer cells, making them suitable for the study and development of anticancer therapies. Three transcription factors were investigated as potential targets in this study. Analysis of string interactions reveals their interaction network. DNA-TF binding was followed by docking with 96 natural compounds to the DNA binding pocket of the transcription factor. Using post-docking processing, compounds were ranked according to their binding energy, hydrogen bond number, and dissociation constant; Withanolide D targeted more than one transcription factor. Therefore, the compound is suitable for in vitro testing using different cancer cell lines.
Thesis
Le récent changement de paradigme lié au développement de la médecine personnalisée en oncologie nécessite une évolution des pratiques impliquant les laboratoires de génétique moléculaire des cancers. Le premier volet de ce travail repose sur les enjeux technologiques de la détection des mutations du gène EGFR pour permettre une prise en charge optimisée des patients éligibles aux thérapies ciblées dans les cancers bronchiques non à petites cellules au diagnostic et lors des récidives. Le séquençage haut débit s’est ici présenté comme la meilleure alternative pour identifier les mutations de sensibilité et de résistance acquise de l’EGFR. Le deuxième volet de cette thèse concerne l’interprétation biologique des altérations génétiques de la tumeur, en particulier les variants des gènes suppresseurs de tumeur BRCA1 et BRCA2 conditionnant la sensibilité aux thérapies ciblées par inhibiteurs de PARP. Deux approches de caractérisation fonctionnelle de variants de signification incertaine (VSI) ont été utilisées, toutes deux réalisables dans un temps compatible avec les impératifs cliniques. La première consiste à caractériser des défauts d’épissage à partir d’ARN tumoral du patient par RT-PCR et clonage rapide des produits de RT-PCR. La deuxième est une évaluation rapide in vitro des VSI par édition du génome en utilisant la technologie CRISPR-Cas9 et un score fonctionnel.
Thesis
p>Mutation detection sensitivity depends on the method employed and the type of mutation present. Melt-MADGE uses a 96-well plate system for loading DNA samples and a combination of temperature ramp and denaturant to resolve both heteroduplexes and mutant homoduplex bands from PCR amplified alleles. Specifically for examining many subjects in parallel, costs will compare very favourably against SSCP, DGGE, dHPLC and direct sequencing. At the time of developing the technique I have designed an assay for mutation scanning for individual amplimers. I had already optimised long and nested PCRs and fine-tuned melt-MADGE assay for 39 amplimers required for the entire coding region of the BRCA1 gene. I have produced high quality results confirming detection of all the common polymorphisms in the largest exon of the gene (exon 11, 3426bp) as well as in other polymorphic exons. In addition, I have discovered a novel polymorphism. In parallel, I have set out to confirm the detection of mutations by an independent mutation detection method, ARMS assay. I have developed ARMS assay for five SNPs in the BRCA1 gene to distinguish the polymorphic samples obtained from the mutated samples. In a pilot study, a panel of 100 anonymous samples from the West Regional Genetics Laboratory (WRGL), were examined. The samples were screened over the entire coding region and we were able to detect several mutations, subsequently confirmed by direct sequencing. Matching the results from the diagnostic laboratory, we achieved 93% sensitivity for mutation detection. Further work is now being directed towards determining the reasons for the reduced sensitivity of the assay in specific cases.</p
Article
Full-text available
The nucleus is an important target of signal transduction by growth factor receptors that stimulate mitogen-activated protein (MAP) kinases. We tested the hypothesis that MAP kinases have a signaling role within the nucleus by examining the effect of the expression of a human MAP kinase isoform (p41mapk) in tissue culture cells. The expressed p41mapk was found to be localized in both the cytoplasmic and nuclear compartments of the cells. Significantly, the expression of p41mapk caused an increase in the phosphorylation of a nuclear substrate: Ser62 of c-Myc. Phosphorylation at Ser62 stimulated the activity of the NH2-terminal transactivation domain of c-Myc. Thus, p41mapk causes the phosphorylation and regulation of a physiologically significant nuclear target of signal transduction. These data establish that at least one MAP kinase isoform has a nuclear role during signal transduction.
Chapter
As progress in cell developmental biology carries on at a breakneck speed, new techniques constantly arise to plug the gaps left by traditional strategies. Cellular Interactions in Development provides detailed discussion and protocols of some of these new techniques, which allow the manipulation of developing organisms such as Drosophila or plants, when and where cells interact with each other to influence their development. The book looks at the really exciting innovations of the identification and functional test of molecules which control these cellular behaviours. The book also describes a number of new ways of hunting for these important proteins involved in cellular communication. A fully comprehensive manual which will prove indispensable to researchers in the fields of cell, developmental, and molecular biology.
Article
Article
Loss of heterozygosity data from familial tumors suggest that BRCA1, a gene that confers susceptibility to ovarian and early-onset breast cancer, encodes a tumor suppressor. The BRCA1 region is also subject to allelic loss in sporadic breast and ovarian cancers, an indication that BRCA1 mutations may occur somatically in these tumors. The BRCA1 coding region was examined for mutations in primary breast and ovarian tumors that show allele loss at the BRCA1 locus. Mutations were detected in 3 of 32 breast and 1 of 12 ovarian carcinomas; all four mutations were germline alterations and occurred in early-onset cancers. These results suggest that mutation of BRCA1 may not be critical in the development of the majority of breast and ovarian cancers that arise in the absence of a mutant germline allele.
Article
According to our present understanding, λ repressor bound to DNA stimulates transcription by touching RNA polymerase bound at an adjacent promoter. The part of repressor required for activation was identified in part by the isolation of mutants specifically impaired in transcriptional activation. The amino acids of repressor altered in these “positive control” mutants lie in an acidic patch on the surface of repressor that is closely apposed to RNA polymerase. In this study, we show that this “activating patch” of repressor is sufficient for transcriptional activation in another sequence context. We transfer this activating patch onto the surface of λ Cro, a protein normally unable to activate transcription, and show that the modified Cro is a transcriptional activator. In addition, we provide evidence that the repressor protein of phage 434 also activates transcription using an activating patch similar to that of λ repressor.
Article
Virion protein 16 (VP16) of herpes simplex virus type 1 contains an acidic transcriptional activation domain. Missense mutations within this domain have provided insights into the structural elements critical for its function. Net negative charge contributed to, but was not sufficient for, transcriptional activation by VP16. A putative amphipathic alpha helix did not appear to be an important structural component of the activation domain. A phenylalanine residue at position 442 was exquisitely sensitive to mutation. Transcriptional activators of several classes contain hydrophobic amino acids arranged in patterns resembling that of VP16. Therefore, the mechanism of transcriptional activation by VP16 and other proteins may involve both ionic and specific hydrophobic interactions with target molecules.