Chromosomal abnormalities of band 8p11 are associated with a distinct subtype of acute myeloid leukemia with French-American-British M4/5 morphology and prominent erythrophagocytosis by the blast cells. This subtype is usually associated with the t(8;16)(p11;p13), a translocation that has recently been shown to result in a fusion between the MOZ and CBP genes. We have cloned the inv(8)(p11q13), an abnormality associated with the same leukemia phenotype, and found a novel fusion between MOZ and the nuclear receptor transcriptional coactivatorTIF2/GRIP-1/NCoA-2. This gene has not previously been implicated in the pathogenesis of leukemia or other malignancies. MOZ-TIF2 retains the histone acetyltransferase homology domains of both proteins and also the CBP binding domain of TIF2. We speculate that the apparently identical leukemia cell phenotype observed in cases with the t(8;16) and the inv(8) arises by recruitment of CBP by MOZ-TIF2, resulting in modulation of the transcriptional activity of target genes by a mechanism involving abnormal histone acetylation.

THE CHARACTERIZATION of recurrent chromosomal abnormalities in leukemia has provided a basis for disease classification and has helped to define prognosis in individual patients. Furthermore, the molecular cloning of the genes affected by these chromosomal changes has considerably advanced our understanding of the mechanisms of leukemogenesis and has enabled the design of molecular approaches to diagnosis and monitoring patient response to treatment.

Translocations involving chromosome band 8p11 are associated with a distinct subtype of acute myeloid leukemia (AML), with blast cells of a monocytoid phenotype that have pronounced erythrophagocytic activity. This subtype is found in approximately 2% of cases of AML with French-American-British M4 or M5 phenotype and typically found in association with the t(8;16)(p11;p13),1 but it is also seen in cases with the t(8;22)(p11;q13), t(8;19)(p11;q13), and inv(8)(p11q13).2-6 Recently the t(8;16) has been cloned and shown to fuse the MOZ gene at 8p11 to the CBP gene at 16p137; the molecular basis of the three other cytogenetic variants is unknown. Translocations of 8p11 are also observed in the 8p11 myeloproliferative syndrome, a rare disorder associated with the t(8;13)(p11;q12), t(8;9)(p11;q32), and t(6;8)(q27;p11).8However, the breakpoints in the t(8;13) are distinct from those associated with de novo AML and do not involve the MOZgene.5 

Both MOZ and CBP have been implicated in histone acetylation,7,9,10 suggesting that the mechanism of leukemogenesis in patients with the t(8;16) involves specific alterations in gene expression by aberrant chromatin remodelling.CBP is also disrupted in the t(11;16), a translocation associated with therapy-related leukemia, by fusion to the MLLgene.11,12 In this paper we show that the inv(8)(p11q13) results in a fusion of MOZ to the nuclear receptor coactivatorTIF2/GRIP-1/NCoA-2 (transcriptional intermediary factor 2, hereafter referred to as TIF2),13-15 and propose a model to explain the similar leukemia phenotypes observed in patients with the t(8;16) and inv(8).

Patient material/details.

Details of the patient's clinical course and leukemia morphology have been described elsewhere.5,6 Briefly, a bone marrow aspirate at presentation was hypercellular, with 98% blasts and prominent erythrophagocytosis; a diagnosis of AML-M5 was made. Cytogenetic analysis revealed an inv(8)(p11q13) in 70% of bone marrow metaphases.

Genomic cloning.

A genomic library was constructed to clone the 7-kb germlineMOZ and 5.1-kb BglII rearranged fragments detected by the MOZ 0.9 probe. Patient bone marrow genomic DNA (0.5 μg) was digested with BglII, ligated to λZAP Express arms and packaged with Gigapack III Gold Packaging Extract according to the manufacturer's instructions (Stratagene, Cambridge, UK). A total of 3.3 × 106plaque-forming units were screened with MOZ 0.9 and five positive clones were recovered. Germline and rearranged bands were distinguished by restriction mapping and one of each was sequenced (model ABI 373A; Applied Biosystems, Foster City, CA).

Fluorescence in situ hybridization (FISH).

A P1 artificial chromosome (PAC) clone (192D10) was isolated by screening the gridded human library RPCI1 (obtained from the Medical Research Council Human Genome Mapping Project (MRC HGMP) Resource Centre, Hinxton, UK) with the probe UN-1. FISH was performed on metaphases from phytohemagglutinin-stimulated peripheral blood lymphocytes from a normal individual. Digoxigenin-labeled probes were prepared by nick translation and detected with sheep antidigoxigenin (Boehringer Mannheim, Lewes, UK), rabbit anti-sheep fluorescein isothiocyanate (FITC; Vector, Bretton, Peterborough, UK), and finally swine anti-rabbit FITC (Dako, High Wycombe, UK). In addition, a biotinylated chromosome 8 centromere dye was included (Oncor, Durham, UK), and detected using Texas Red-Avidin (Vector). Chromosomes were counterstained with DAPI/antifade (Biovation, Aberdeen, UK). FISH-labeled metaphases were examined using an Olympus Vanox microscope, equipped with a fluorescence unit, a charge-coupled device camera, and images captured on SmartCapture Software (Vysis, Richmond, UK).

Rapid amplification of cDNA ends (RACE)–polymerase chain reaction (PCR).

One microgram of total patient bone marrow RNA was reverse-transcribed with primer RCT-A (5′-ATTGGTAGCTCTTGATCNNNNNN-3′) using standard conditions.16 A portion of cDNA was amplified with MOZ-RCO (5′-CCCTAGAGAATACTTCCGTC-3′) and a primer made up of the unique sequence from RCT-A (RC-B; 5′-ATTGGTAGCTCTTGATC-3′). Hemi-nested amplification was performed with an inner MOZ primer (MOZ-RCI: 5′-GGATGTACTCAGGTGTCAGT-3′) and RC-B. PCR products were shotgun cloned into pCR2.1 (Invitrogen, Leeu, The Netherlands), screened with a MOZ probe, and sequenced.

Yeast artificial chromosome (YAC) clones.

A gridded CEPH mega YAC library (obtained from the MRC HGMP Resource Centre) was screened with probes for the 5′ and 3′ ends of the published TIF2 cDNA sequence.13 Probes were amplified from human genomic DNA using the primers TIF2-A (5′-GGCACAGTTGCTGATATGTG-3′) and TIF2-B (5′-GTCCAAGTTGGTCAGGACAT-3′) for the 5′ end of TIF2, and TIF2-C (5′-GTGGCCTGCTTAGTAACATG-3′) and TIF2-D (5′-GGCTTGATACCAATCGAGCT-3′) for the 3′ end. The positions of positive clones were compared with existing chromosome 8 contig maps.17,18 Confirmation that these clones contained the TIF2 gene was obtained by PCR using the same primers combinations.

Southern blotting of patient bone marrow DNA with a MOZcDNA probe, MOZ 0.9, that spanned the published t(8;16) breakpoints revealed normal- and abnormal-sized bands after digestion withBglII5 (Fig 1A). A bacteriophage λ library was constructed from BglII-digested patient DNA and screened with MOZ 0.9. Two types of clone were recovered, corresponding to the normal 7-kb and rearranged 5.1-kb bands, respectively.

Fig. 1.

(A) Southern Blot analysis. Genomic DNA from four controls (lanes 1-4) and the inv(8) patient (lane 5) were digested withBglII (i-iii) or EcoRI (iv-vi) and probed with MOZ 0.9, MOZ I, and UN-1 as indicated. Germline bands were present in all samples and an additional band was seen for the patient for all enzyme/probe combinations except EcoRI/MOZ 0.9. (B) Schematic representation of the two sequenced bacteriophage λ clones. The 7-kb and 5.1-kb BglII fragments were derived from the germline and rearranged bands, respectively. The last MOZ exon is shown as a filled box and intron sequence as a line. The positions of the probes MOZ 0.9, MOZ I, and UN-1 are indicated. The breakpoint in the 5.1-kb clone coincides with the EcoRI site within the last exon of MOZ, 47 bp downstream of the MOZ intron-exon junction.

Fig. 1.

(A) Southern Blot analysis. Genomic DNA from four controls (lanes 1-4) and the inv(8) patient (lane 5) were digested withBglII (i-iii) or EcoRI (iv-vi) and probed with MOZ 0.9, MOZ I, and UN-1 as indicated. Germline bands were present in all samples and an additional band was seen for the patient for all enzyme/probe combinations except EcoRI/MOZ 0.9. (B) Schematic representation of the two sequenced bacteriophage λ clones. The 7-kb and 5.1-kb BglII fragments were derived from the germline and rearranged bands, respectively. The last MOZ exon is shown as a filled box and intron sequence as a line. The positions of the probes MOZ 0.9, MOZ I, and UN-1 are indicated. The breakpoint in the 5.1-kb clone coincides with the EcoRI site within the last exon of MOZ, 47 bp downstream of the MOZ intron-exon junction.

Close modal

Sequence analysis of a 7-kb clone revealed a single large MOZexon of 4 kb from position 3746 of the cDNA (Genbank: U47742) to theBglII site at position 7742. Sequence of a 5.1-kb clone showed a break near the beginning of this exon within the coding sequence (position 3793), with 1.1 kb of unique sequence fused toMOZ (Fig 1B). The single published MOZ breakpoint in a patient with a t(8;16) also interrupted the same exon, but there was no sequence homology between that breakpoint and the one found here. A probe derived from the 1.1 kb of unique sequence (UN-1) was used to isolate a PAC clone which, as expected, mapped to 8q13 as determined by FISH (Fig 2).

Fig. 2.

FISH. PAC clone 192D10 was hybridized to normal lymphocyte metaphases along with a chromosome 8 centromere probe. Hybridization of the PAC is seen at 8q13.

Fig. 2.

FISH. PAC clone 192D10 was hybridized to normal lymphocyte metaphases along with a chromosome 8 centromere probe. Hybridization of the PAC is seen at 8q13.

Close modal

Primers were designed from the MOZ cDNA sequence upstream of the last exon and used for 3′ RACE-PCR with RNA extracted from the patient's bone marrow. In addition to normal MOZ products, clones were isolated in which the penultimate exon of this gene was fused to novel sequence. A search of the Genbank database showed that the sequence was derived from the TIF2 gene, a recently described member of the nuclear receptor transcriptional coactivator family of proteins.13-15 Nucleotide 3745 ofMOZ was joined to nucleotide 2768 of TIF2 (Genbank:X97674), resulting in an in-frame fusion (Fig3A).

Fig. 3.

(A) Sequence surrounding the MOZ-TIF2 fusion. Nucleotide 3745 of MOZ was joined to nucleotide 2768 ofTIF2. The underlined sequence indicates the region withinTIF2 that is alternatively spliced in both the normal and fusion genes. (B) RT-PCR analysis of bone marrow RNA from a normal individual and the patient with the inv(8). Normal MOZ andTIF2 transcripts were detected in both samples, the lowerTIF2 band resulting from alternative splicing. TheMOZ-TIF2 fusion was seen only in the patient and reciprocalTIF2-MOZ transcripts were not detected. The identity of all bands was confirmed by sequencing. M, 123-bp molecular weight marker.

Fig. 3.

(A) Sequence surrounding the MOZ-TIF2 fusion. Nucleotide 3745 of MOZ was joined to nucleotide 2768 ofTIF2. The underlined sequence indicates the region withinTIF2 that is alternatively spliced in both the normal and fusion genes. (B) RT-PCR analysis of bone marrow RNA from a normal individual and the patient with the inv(8). Normal MOZ andTIF2 transcripts were detected in both samples, the lowerTIF2 band resulting from alternative splicing. TheMOZ-TIF2 fusion was seen only in the patient and reciprocalTIF2-MOZ transcripts were not detected. The identity of all bands was confirmed by sequencing. M, 123-bp molecular weight marker.

Close modal

Because the chromosomal localization of TIF2 had not previously been determined, we hybridized probes generated from the 5′ and 3′ ends of this gene to a gridded CEPH megaYAC library. Several positive clones were isolated that contained the entire TIF2 gene, including the YACs 798c3 and 815d1 which have been mapped previously to 8q13.2-3.17 

The presence of a novel fusion transcript was confirmed by reverse transcriptase-PCR using cDNA primers either side of the junction. MOZ-TIF2 was specifically amplified from the patient's bone marrow RNA, but not from bone marrow RNA extracted from normal individuals (Fig 3B). Reciprocal TIF2-MOZ transcripts were not detected. Two MOZ-TIF2 bands were amplified, which, by sequencing, were found to result from alternative splicing within theTIF2 moiety (nucleotides 2767-2975). The same alternative splice was found for TIF2 amplified from both the patient and normal individual. Rearrangement of the TIF2 gene was confirmed by Southern blotting. After digestion with BglII, probe UN-1 detected the same-sized rearranged band as MOZ 0.9 (Fig 1A). Rearrangements were not seen in digests of DNA from normal individuals.

The MOZ gene was isolated as a consequence of its fusion toCBP by the t(8;16) in AML. Of its structural features, the most significant are a PHD/LAP domain, thought to be involved in protein-protein interactions,19,20 and a region that is homologous to a number of lower eukaryotic histone acetyltransferases (HATs).7 Although the precise function of MOZ is not known, a related gene in Drosophila, MOF, is required for dosage compensation, the mechanism which enables male flies with a single X chromosome to express the same level of X-linked products as females with two X chromosomes.21 The related yeast genesSAS2 and YBF2 influence transcriptional silencing and, taken together, it is likely that MOZ is a chromatin-bound HAT that modulates the transcription of specific target genes. CBP is a cointegrator/adaptor that is believed to coordinate the transcriptional effects of multiple signals from cell surface and nuclear receptors.22-24 Recently it has been shown that CBP also has HAT activity.9,10 

Nuclear receptors are ligand-inducible transcription factors, which typically consist of three structural domains: an N-terminus containing an activation function, AF-1; a DNA-binding domain; and a ligand-binding C-terminus that contains a second activation function, AF-2.25 AF-2 mediates transcriptional activation through nuclear receptor coactivators (NRCoAs),26,27 which are thought to stimulate gene expression by facilitating the assembly of basal transcription factors into a stable preinitiation complex.28 Transcriptional interference/squelching experiments have indicated that NRCoAs are limiting factors in this process.13,TIF2 was recently shown to be one such mediator of AF-2 function.13 This protein is homologous to other NRCoAs, specifically SRC-1 (also known as F-SRC-1 and NCoA-1),15,29,30 and pCIP (also known as RAC3, ACTR, and AIB1).15,31-33 Although the functional domains of TIF2 have not been determined, the related proteins SRC-1/F-SRC-1/NCoA-1 and ACTR/pCIP/RAC3/AIB1 have HAT activity and also interact directly with CBP.29,34-36 Therefore, it is likely that NRCoAs mediate transcriptional activation by a mechanism involving extensive chromatin remodeling. Acetylation of amino termini of core histones allows nucleosomes to unfold, thus increasing access to transcription factors.37 Conversely, histone deacetylation has been shown to repress transcription, at least in some situations.38 

The MOZ-TIF2 fusion retains the LAP finger and HAT homology domains of MOZ, along with the putative CBP interacting domain (CID) and HAT domain of TIF2 (Fig 4). The fusion does not retain the TIF2 PAS/bHLH domain, believed to be involved in DNA binding and protein heterodimerization, or the receptor interacting domain (RID), which mediates the binding of transcriptional coactivators to nuclear receptors via conserved LXXLL motifs.15,29,39Therefore, it is unlikely that MOZ-TIF2 is able to interact with any of the upstream components that normally require TIF2 as a transcriptional intermediary. Instead, the HAT activity of TIF2 may directly modulate or augment the transcriptional activity of genes normally regulated by MOZ (Fig 5A). Alternatively, the TIF2 moiety may serve as a bridge between MOZ and CBP, and it is the HAT or other activities of CBP that are relevant to putative leukemogenic alterations in gene expression (Fig 5B). The latter model is particularly attractive in view of the striking similarity in cell phenotypes associated with the MOZ-CBP fusion in the t(8;16) (Fig 5C) and MOZ-TIF2 in the inv(8). The molecular basis of the two variant translocations t(8;19) and t(8;22) is unknown, but they may result in analogous mechanisms of leukemogenesis. For example, it has been postulated that the t(8;22) may involve p300 at 22q13, a gene that is structurally and functionally closely related toCBP.7 Indeed, it has been shown recently thatp300 is fused to the MLL gene in AML with the t(11;22)(q23;q13).40 

Fig. 4.

Schematic representation of MOZ, TIF2, and MOZ-TIF2 fusion proteins. Domains are indicated as follows: LAP, leukemia-associated protein20; HAT, histone acetyltransferase; M-rich, methionine-rich; CID and RID, putative CBP and nuclear receptor interacting domains based on homology with SRC-1.29,30 The MOZ-TIF2 fusion retains the LAP finger and HAT homology domains of MOZ, along with the CID and HAT domains of TIF2.

Fig. 4.

Schematic representation of MOZ, TIF2, and MOZ-TIF2 fusion proteins. Domains are indicated as follows: LAP, leukemia-associated protein20; HAT, histone acetyltransferase; M-rich, methionine-rich; CID and RID, putative CBP and nuclear receptor interacting domains based on homology with SRC-1.29,30 The MOZ-TIF2 fusion retains the LAP finger and HAT homology domains of MOZ, along with the CID and HAT domains of TIF2.

Close modal
Fig. 5.

Hypothetical models of the mode of action of the MOZ-TIF2 fusion protein. (A) TIF2 may directly modulate the transcriptional activity of genes normally regulated by MOZ through the addition or removal of histone acetyl (Ac−) groups by its histone acetyltransferase (HAT) domain. (B) The TIF2 moiety may serve as a bridge between MOZ and CBP, and it is the HAT or other activities of CBP that leads to leukemogenic alterations in gene expression. Chromatin-associated CBP may be responsive to other cellular signals such as those mediated by jun, CREB, or STAT proteins.22-24(C) The MOZ-CBP fusion in the t(8;16),7 which is associated with a strikingly similar leukemia cell phenotype to that seen in cases with the inv(8).

Fig. 5.

Hypothetical models of the mode of action of the MOZ-TIF2 fusion protein. (A) TIF2 may directly modulate the transcriptional activity of genes normally regulated by MOZ through the addition or removal of histone acetyl (Ac−) groups by its histone acetyltransferase (HAT) domain. (B) The TIF2 moiety may serve as a bridge between MOZ and CBP, and it is the HAT or other activities of CBP that leads to leukemogenic alterations in gene expression. Chromatin-associated CBP may be responsive to other cellular signals such as those mediated by jun, CREB, or STAT proteins.22-24(C) The MOZ-CBP fusion in the t(8;16),7 which is associated with a strikingly similar leukemia cell phenotype to that seen in cases with the inv(8).

Close modal

Although TIF2 has not previously been implicated in malignancy, several sporadic leukemias have been described with translocations or other structural rearrangements that involve chromosome band 8q13.41 Furthermore, the related geneAIB1/pCIP/RAC3/ACTR at 20q12 is amplified in breast and ovarian cancers,33 and, in addition, the ARA70gene was fused to RET in a case of human thyroid papillary carcinoma42,43 and TIF1 fused to B-RAF in the mouse hepatoma-derived oncogene T18.44 Although ARA70 and TIF1 do not share sequence homology with the TIF2 family of NRCoAs, they are also involved in the ligand-dependent activation function of nuclear receptors.43,44 These observations suggest that subversion of the mechanisms by which nuclear receptors modulate gene transcription, including specific acetylation of histones, is widely involved in malignancy.

We thank the MRC HGMP Resource Centre for providing the PAC clones and CEPH megaYAC clones.

Supported by the Leukaemia Research Fund. R.C.T.A. was supported by CNPq (Conselho Nacional de Desenvolvimento Cientı́fico e Tecnológico-No 200995/94-4), Brazil.

Address reprint requests to Nicholas C.P. Cross, PhD, Dept of Haematology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Rd, London, W12 ONN, UK.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked "advertisement" is accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Mitelman
F
Heim
S
Quantitative acute leukemia cytogenetics.
Genes Chromosom Cancer
5
1992
57
2
Heim
S
Avanzi
GC
Billström
R
Kristoffersson
U
Mandahl
N
Bekassy
AN
Garwicz
S
Wiebe
T
Pegoraro
L
Falda
M
Resegotti
L
Mitelman
F
A new specific chromosomal rearrangement, t(8;16)(p11;p13), in acute monocytic leukaemia.
Br J Haematol
66
1987
323
3
Laı̈
JL
Zandecki
M
Jouet
JP
Savary
JB
Lambiliotte
A
Bauters
F
Cosson
A
Deminatti
M
Three cases of translocation (8;16)(p11;p13) observed in acute myelomonocytic leukemia: A new specific subgroup?
Cancer Genet Cytogenet
27
1987
101
4
Brizard
A
Guilhot
F
Huret
JL
Benz-Lemoine
E
Tanzer
J
The 8p11 anomaly in “monoblastic” leukaemia.
Leuk Res
12
1988
693
5
Aguiar
RCT
Chase
A
Coulthard
S
Macdonald
DHC
Carapeti
M
Reiter
A
Sohal
J
Lennard
A
Goldman
JM
Cross
NCP
Abnormalities of chromosome band 8p11 in leukemia: Two clinical syndromes can be distinguished on the basis of MOZ involvement.
Blood
90
1997
3130
6
Coulthard
S
Chase
A
Orchard
K
Watmore
A
Vora
A
Goldman
JM
Swirsky
DM
Two cases of inv(8)(p11q13) in AML with erythrophagocytosis; a new cytogenetic variant.
Br J Haematol
100
1998
561
7
Borrow
J
Stanton VP Jr
Andresen
JM
Becher
R
Behm
FG
Chaganti
RSK
Civin
CI
Disteche
C
Dubé
I
Frischauf
AM
Horsman
D
Mitelman
F
Volinia
S
Watmore
AE
Housman
DE
The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein.
Nat Genet
14
1996
33
8
Macdonald
DHC
Aguiar
RCT
Mason
PJ
Goldman
JM
Cross
NCP
A new myeloproliferative disorder associated with chromosomal translocations involving 8p11: A review.
Leukemia
9
1995
1628
9
Bannister
AJ
Kouzarides
T
The CBP co-activator is a histone acetyltransferase.
Nature
384
1996
641
10
Ogryzko
VV
Schiltz
RL
Russanova
V
Howard
B
Nakatani
Y
The transcriptional coactivators p300 and CBP are histone acetyltransferases.
Cell
87
1996
953
11
Rowley
JD
Reshmi
S
Sobulo
O
Musvee
T
Anastasi
J
Raimondi
S
Schneider
NR
Barredo
JC
Cantu
ES
Schlegelberger
B
Behm
F
Doggett
NA
Borrow
J
Zeleznik-Le
N
All patients with the t(11;16)(q23;p13.3) that involves MLL and CBP have treatment-related hematologic disorders.
Blood
90
1997
535
12
Taki
T
Sako
M
Tsuchida
M
Hayashi
Y
The t(11;16)(q23;p13) translocation in myelodysplastic syndrome fuses the MLL gene to the CBP gene.
Blood
89
1997
3945
13
Voegel
JJ
Heine
MJS
Zechel
C
Chambon
P
Gronemeyer
H
TIF2, a 160kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors.
EMBO J
15
1996
3667
14
Hong
H
Kohli
K
Trivedi
A
Johnson
DL
Stallcup
MR
GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors.
Proc Natl Acad Sci USA
93
1996
4948
15
Torchia
J
Rose
DW
Inostroza
J
Kamei
Y
Westin
S
Glass
CK
Rosenfeld
MG
The transcriptional co-activator p/CIP binds CBP and mediates nuclear-receptor function.
Nature
387
1997
677
16
Cross
NCP
Hughes
TP
Lin
F
O'Shea
P
Bungey
J
Marks
DI
Ferrant
A
Martiat
P
Goldman
JM
Minimal residual disease after allogeneic bone marrow transplantation for chronic myeloid leukaemia in first chronic phase: Correlations with acute graft-versus-host disease and relapse.
Br J Haematol
84
1993
67
17
Chumakov IM, Rigault P, Le Gall I, Bellanné-Chantelot C, Billault A, Guillou S, Soularue P, Guasconi G, Poullier E, Gros I, Belova M, Sambucy J-L, Susini L, Gervy P, Gilbert F, Beaufils S, Bui H, Massart C, De T and M-F, Dukasz F, Lecoulant S, Ougen P, Perrot V, Saumier M, Soravito C, Bahouayila R, Cohen-Akenine A, Barillot E, Bertrand S, Codani J-J, Caterina D, Georges I, Lacroix B, Lucotte G, Sahbatou M, Schmit C, Sangouard M, Tubacher E, Dib C, Fauré S, Fizames C, Gyapay G, Millasseau P, Nguyen S, Muselet D, Vignal A, Morissette J, Menninger J, Lieman J, Desai T, Banks A, Bray-Ward P, Ward D, Hudson T, Gerety S, Foote S, Stein L, Page DC, Lander ES, Weissenbach J, Le Paslier D, Cohen D: A YAC contig map of the human genome. Nature 377:175, 1995
18
Dib
C
Faure
S
Fizames
C
Samson
D
Drouot
N
Vignal
A
Millasseau
P
Marc
S
Hazan
J
Seboun
E
Lathrop
M
Gyapay
G
Morissette
J
Weissenbach
J
A comprehensive genetic map of the human genome based on 5,264 microsatellites.
Nature
380
1996
152
19
Aasland
R
Gibson
TJ
Steward
AF
The PHD finger: Implications for chromatin-mediated transcriptional regulation.
Trends Biochem Sci
20
1995
56
20
Saha
V
Chaplin
T
Gregorini
A
Ayton
P
Young
BD
The leukemia-associated-protein (LAP) domain, a cysteine-rich motif, is present in a wide range of proteins, including MLL, AF10, and MLLT6 proteins.
Proc Natl Acad Sci USA
92
1995
9737
21
Hilfiker
A
Hilfiker-Kleiner
D
Pannuti
A
Lucchesi
JC
mof, a putative acetyl transferase gene related to the Tip60 and MOZ human genes and to the SAS genes of yeast, is required for dosage compensation in Drosophila.
EMBO J
16
1997
2054
22
Kwok
RPS
Lundblad
JR
Chrivia
JC
Richards
JP
Bächinger
HP
Brennan
RG
Roberts
SGE
Green
MR
Goodman
RH
Nuclear protein CBP is a coactivator for the transcription factor CREB.
Nature
370
1994
223
23
Chakravarti
D
LaMorte
VJ
Nelson
MC
Nakajima
T
Schulman
IG
Juguilon
H
Montminy
M
Evans
RM
Role of CBP/p300 in nuclear receptor signalling.
Nature
383
1996
99
24
Kamei
Y
Xu
L
Heinzel
T
Torchia
J
Kurokawa
R
Gloss
B
Lin
S-C
Heyman
RA
Rose
DW
Glass
CK
Rosenfeld
MG
A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors.
Cell
85
1996
403
25
Evans
RM
The steroid and thyroid hormone receptor superfamily.
Science
240
1988
889
26
Horwitz
KB
Jackson
TA
Bain
DL
Richer
JK
Takimoto
GS
Tung
L
Nuclear receptor coactivators and corepressors.
Mol Endocrinol
10
1996
1167
27
Glass
CK
Rose
DW
Rosenfeld
MG
Nuclear receptor coactivators.
Curr Opin Cell Biol
9
1997
222
28
Klein-Hitpass
L
Tsai
SY
Weigel
NL
Allan
GF
Riley
D
Rodriguez
R
Schrader
WT
Tsai
M-J
O'Malley
BW
The progesterone receptor stimulates cell-free transcription by enhancing the formation of a stable preinitiation complex.
Cell
60
1990
247
29
Oñate
SA
Tsai
SY
Tsai
M-J
O'Malley
BW
Sequence and characterization of a coactivator for the steroid hormone receptor superfamily.
Science
270
1995
1354
30
Takeshita
A
Yen
PM
Misiti
S
Cardona
GR
Liu
Y
Chin
WW
Molecular cloning and properties of a full-length putative thyroid hormone receptor coactivator.
Endocrinology
137
1996
3594
31
Li
H
Gomes
PJ
Chen
JD
RAC3, a steroid/nuclear receptor-associated coactivator that is related to SRC-1 and TIF2.
Proc Natl Acad Sci USA
94
1997
8479
32
Chen
H
Lin
RJ
Schiltz
RL
Chakravarti
D
Nash
A
Nagy
L
Privalsky
ML
Nakatani
Y
Evans
RM
Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300.
Cell
90
1997
569
33
Anzick
SL
Kononen
J
Walker
RL
Azorsa
DO
Tanner
MM
Guan
X-Y
Sauter
G
Kallioniemi
O-P
Trent
JM
Meltzer
PS
AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer.
Science
277
1997
965
34
Smith
CL
Oñate
SA
Tsai
M-J
O'Malley
BW
CREB binding protein acts synergistically with steroid receptor coactivator-1 to enhance steroid receptor-dependent transcription.
Proc Natl Acad Sci USA
93
1996
8884
35
Yao
T-P
Ku
G
Zhou
N
Scully
R
Livingston
DM
The nuclear hormone receptor coactivator SRC-1 is a specific target of p300.
Proc Natl Acad Sci USA
93
1996
10626
36
Spencer
TE
Jenster
G
Burcin
MM
Allis
CD
Zhou
J
Mizzen
CA
McKenna
NJ
Oñate
SA
Tsai
S
Tsai
M-J
O'Malley
BW
Steroid receptor coactivator-1 is a histone acetyltransferase.
Nature
389
1997
194
37
Grunstein
M
Histone acetylation in chromatin structure and transcription.
Nature
389
1997
349
38
Pazin
MJ
Kadonaga
JT
What's up and down with histone deacetylation and transcription?
Cell
89
1997
325
39
Heery
DM
Kalkhoven
E
Hoare
S
Parker
MG
A signature motif in transcriptional co-activators mediates binding to nuclear receptors.
Nature
387
1997
733
40
Ida
K
Kitabayashi
I
Taki
T
Taniwaki
M
Noro
K
Yamamoto
M
Ohki
M
Hayashi
Y
Adenoviral E1A-associated protein p300 is involved in acute myeloid leukemia with t(11;22)(q23;q13).
Blood
90
1997
4699
41
Mitelman F: Catalog of Chromosome Aberrations in Cancer. New York, NY, Wiley, 1991
42
Santoro
M
Dathan
NA
Berlingieri
MT
Bongarzone
I
Paulin
C
Grieco
M
Pierotti
MA
Vecchio
G
Fusco
A
Molecular characterization of RET/PTC3; a novel rearranged version of the RET proto-oncogene in a human thyroid papillary carcinoma.
Oncogene
9
1994
509
43
Yeh
S
Chang
C
Cloning and characterization of a specific coactivator, ARA70, for the androgen receptor in human prostate cells.
Proc Natl Acad Sci USA
93
1996
5517
44
Le Douarin
B
Zechel
C
Garnier
J-M
Lutz
Y
Tora
L
Pierrat
B
Heery
D
Gronemeyer
H
Chambon
P
Losson
R
The N-terminal part of TIF1, a putative mediator of the ligand-dependent activation function (AF-2) of nuclear receptors, is fused to B-raf in the oncogenic protein T18.
EMBO J
14
1995
2020
Sign in via your Institution