Skip to main content
Erschienen in: Cancer and Metastasis Reviews 4/2014

Open Access 01.12.2014

Towards an understanding of the structure and function of MTA1

verfasst von: Christopher J. Millard, Louise Fairall, John W. R. Schwabe

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 4/2014

Abstract

Gene expression is controlled through the recruitment of large coregulator complexes to specific gene loci to regulate chromatin structure by modifying epigenetic marks on DNA and histones. Metastasis-associated protein 1 (MTA1) is an essential component of the nucleosome remodelling and deacetylase (NuRD) complex that acts as a scaffold protein to assemble enzymatic activity and nucleosome targeting proteins. MTA1 consists of four characterised domains, a number of interaction motifs, and regions that are predicted to be intrinsically disordered. The ELM2-SANT domain is one of the best-characterised regions of MTA1, which recruits histone deacetylase 1 (HDAC1) and activates the enzyme in the presence of inositol phosphate. MTA1 is highly upregulated in several types of aggressive tumours and is therefore a possible target for cancer therapy. In this review, we summarise the structure and function of the four domains of MTA1 and discuss the possible functions of less well-characterised regions of the protein.

1 Introduction

The class I histone deacetylase (HDAC) corepressor complexes are multi-protein assemblies that contain one or more enzymes that modify chromatin, components with nucleosome or DNA binding activity and proteins that function to provide a scaffold to the complex. Five major HDAC-containing corepressor complexes have been described and include nucleosome remodelling and deacetylase (NuRD) [1, 2], COREST [3], Sin3A [4, 5], MIDAC [6] and NCoR/SMRT [4, 7]. Understanding the distinct features and architectures of these complexes is likely to be important for the design of specifically targeted therapeutics.
Metastasis-associated protein 1 (MTA1) and its homologues MTA2 and MTA3 are essential components of the NuRD corepressor complex (reviewed in [810]). The MTA proteins do not have any intrinsic enzymatic activity but play a key role in stabilising and assembling the complex. MTA1 was first isolated from a metastatic breast cancer cell line and has since been shown to be upregulated in several other metastatic human cancers [1113]. Elevated expression levels of MTA1 are strongly associated with the growth of aggressive endometrial, breast and ovarian cancers, and can be used as a prognosis marker in the progression of human cancers [1416].
Over half of the residues of MTA1 are predicted to be intrinsically disordered, and this has presented a major challenge in the structural characterisation of this protein (Fig. 1). However, significant progress has been made in studying domains of MTA1 in complex with other proteins from the NuRD complex. In this review, we examine the structural insights that have been gained for MTA1, MTA2 and MTA3. We also discuss domains and motifs of MTA1 that have not been structurally characterised, but for which structural information is available from related proteins.

2 The NuRD corepressor complex

The NuRD corepressor complex consists of six protein subunits: HDAC1/2, MTA1/2/3, p66 α/β, histone-binding protein  4/7 (RBBP4/7), methyl-CpG-binding protein 2/3 (MBD2/3) and chromatin-helicase-DNA-binding protein 3/4 (CHD3/4). Deacetylase and helicase activities are combined in this complex. HDAC1 and HDAC2 are single domain deacetylase enzymes that remove the acetyl group from modified lysines resulting in an extra positive charge on histone tails. This leads to a more compact chromatin structure as well as removing binding sites for proteins that contain bromodomains [17]. CHD3 and CHD4 contain subunits with ATP-helicase activity that remodels nucleosomes [18, 19]. This activity dynamically controls the accessibility of chromatin to DNA binding proteins and additional coregulators.
Other proteins within the core NuRD complex support and enhance these enzyme activities, as well as facilitating recruitment to chromatin. RBBP4 and RBBP7 (RbAp48/46) are ∼50 kDa WD40 domain proteins that interact with chromatin and have been shown to bind to histone H4 tails [20]. p66α and p66β (GATAD2A/B) contain GATA zinc finger domains and are involved in binding to unmodified histone tails [21]. MBD2 and MBD3 have been shown to bind to methylated cytosine-guanosine dinucleotides (CpGs) [22]. MBD2 has been found to localise at transcriptional start sites with methylated CpG islands, and this enrichment coincides with gene repression. MTA1 and the closely related MTA2 and MTA3 proteins enhance and direct the activity of HDAC1 to modify chromatin [1, 23]. MTA1 has been shown to directly interact with RBBP4, HDAC1 and CHD4 within the NuRD complex [2427].

3 MTA family proteins

MTA1 is an 80-kDa protein that has four conserved domains within the amino-terminal half of the protein. These are a bromo-adjacent homology (BAH) domain; an egl-27 and MTA1 homology domain 2 (ELM2); a Swi3, Ada2, NCoR and TFIIIB domain (SANT); and GATA-zinc finger domain (GATA) (Fig. 1). MTA1 and its homologues MTA2 and MTA3 share 63 and 72 % identity, respectively. The proteins are essentially identical within these four structured domains with the only notable difference being an extended loop within the MTA1-BAH domain. MTA1 is the largest protein in the family with 715 residues; MTA2 has 668 residues; and MTA3 has 594 residues. The carboxy-termini are significantly more divergent, which may explain why MTA1, MTA2 and MTA3 are found in mutually exclusive complexes and are implicated in different signalling pathways [14].
MTA1 can be differentially spliced, and the four conserved domains are truncated or missing in the resulting isoforms [28]. MTA1-short (MTA1s) is one of the best-characterised splice variants, being around half the size of full-length MTA1 and is truncated between the SANT and GATA domain. The original carboxy-terminus is replaced with a 33-residue extension containing the nuclear receptor box motif (LxxLL). The LxxLL motif interacts with the AF2 domain of ERα and results in the localisation of ERα to the cytoplasm in estrogen-positive breast cancer cells [29]. Another isoform known as ZG29p contains just the carboxy-terminus of MTA1 and is encoded by the last seven exons [30]. The resulting protein is missing all four conserved domains. A number of further isoforms have been characterised and confirm that the carboxy-terminus must contain at least one nuclear localisation signal for MTA1 to be retained within to the nucleus [28].
In addition to these four conserved domains, MTA1 contains two motifs that have the potential to promote SH3-binding and five SPXX motifs that are highly conserved across species [3133]. Although these MTA1 motifs are yet to be fully characterised, considerable insight into the structure and function of these domains can be gained through sequence analysis. Comparison with related protein families can be used to better understand the assembly and function of the core NuRD corepressor complex.

4 Structurally characterised domains of the MTA family

There are an increasing number of structures of MTA family domains in isolation and in complex with binding partners. The first structural information for the MTA proteins was a structure of the SANT domain from MTA3 that was deposited in the protein databank (PDB code 2CRG) but without any corresponding publication. More recently, Millard et al. determined the structure of the ELM2 and SANT domains from MTA1 bound to HDAC1, revealing the nature of the interface between the two proteins as well as highlighting a mechanism of regulation through inositol phosphate signalling [26]. Finally, a carboxy-terminal fragment of MTA1 has been crystallised bound to RBBP4 (RbAp48) [25]. Insights from the structural as well as functional studies of these domains are discussed below.

4.1 The SANT domain and HDAC activation by inositol phosphates

The SANT domain is a relatively small domain of around 50 amino acids and is found in a number of proteins that are involved in transcription regulation [34]. The domain was originally identified in the nuclear receptor corepressor protein NCOR1 and in chromatin modifying proteins SWI3 and ADA2 [35]. The domain folds to form a three-helix bundle around a small hydrophobic core and is highly similar to the DNA-binding domain of Myb-related proteins.
The NMR structure of the MTA3-SANT domain confirmed the predicted helix-turn-helix arrangement (Fig. 2). Inspection of the structure suggests that the SANT domain is functionally divergent from the Myb DNA-binding domain [36]. Important basic DNA recognition residues within helix 3 (or the recognition helix) are not conserved, suggesting that the SANT domain would be unable to make sequence-specific contacts to DNA. In addition, the SANT domain contains three bulky hydrophobic residues at the carboxy-terminus of helix 3 that would sterically hinder DNA binding. This suggests that, despite the conserved fold, these domains have evolved to perform different functions.
More recently, the structure of a longer fragment of MTA1 that includes both the ELM2 and SANT domains bound to HDAC1 has been solved by X-ray crystallography. This structure shows that helix 3 of the SANT domain forms a basic pocket at the interface between HDAC1 and MTA1, near the active site of the enzyme [26]. This pocket has implications for enzyme activation of class I HDACs (see below).
Sequence alignments show that the SANT domain of MTA1 is highly related to the first of two SANT domains from the nuclear receptor corepressor protein SMRT (also known as NCOR2). The SMRT-SANT is a critical partner of HDAC3 and is essential for HDAC3 activity [37]. The structure of HDAC3 bound to SMRT-SANT has been solved and shows that a inositol-(1,4,5,6)-tetrakisphosphate (IP4) molecule can bind into a basic pocket at the interface between the two proteins [38]. Importantly, IP4 has a key role in activating the enzyme, since high salt treatment causes HDAC3 to become inactive and adding back exogenous IP4 leads to enhanced HDAC activity. The concentration of IP4 required for activation of HDAC3 is comparable to the measured levels of inositol phosphates in the cell [39].
The residues coordinating IP4 binding in HDAC3 and SMRT are conserved in HDAC1 and MTA1-SANT, and a similar basic pocket is present at the interface between the two proteins. This pocket is formed through the contribution of a lysine and two arginine residues from HDAC1, and two lysine and two tyrosine residues from MTA1. The similarity in size and charge of the pocket suggested that an inositol phosphate molecule could be accommodated. However, in the crystal structure, an inositol phosphate molecule is not present. Native mass spectroscopy showed that the inositol phosphate is lost during purification and is replaced during crystallisation by ordered sulphate molecules (2 M ammonium sulphate was used as a precipitant). As with HDAC3:SMRT, the addition of IP4 to HDAC1:MTA1 enhances HDAC activity, and mutation of the IP4 coordinating residues inhibits activation [26].
Interestingly, the inositol phosphate interacting residues are not only conserved in SMRT-SANT and MTA-SANT domains but also in related corepressor proteins such as RCOR1-3, MIER1-3 and RERE. These SANT domain-containing proteins are recruited to distinct corepressor complexes that recruit HDAC1, and all could potentially form an inositol-binding pocket. Since HDAC1 and HDAC2 are highly homologous, the binding of inositol phosphates will be a common activating mechanism for HDAC1, 2 and 3 (i.e. all class I HDAC corepressor complexes).

4.2 The ELM2 domain and dimerisation of the NuRD complex

The ELM2 domain within corepressor proteins such as MTA1-3, RCOR1-3, MIER1-3 and RERE has been shown to be required for the recruitment of HDAC1 and HDAC2 to the respective corepressor complexes. The ELM2 domain is positioned immediately amino-terminal to the SANT domain. Structure prediction of the MTA1-ELM2 domain suggests that the domain is largely intrinsically disordered (i.e. lacking an intrinsically fixed structure), although there are predicted helical regions at the carboxy-terminus. Circular dichroism experiments show that the ELM2 domain contains essentially no secondary structure when expressed in isolation [26]. However, when coexpressed with HDAC1, the ELM2-SANT domain forms a stable complex with the HDAC enzyme [26].
The crystal structure of the complex between HDAC1 and the ELM2-SANT domain from MTA1 shows that the ELM2 domain is divided into two structural regions [26]. The amino-terminal part of the ELM2 domain adopts an extended conformation that wraps around the HDAC making multiple interactions. Close to the amino-terminus, there is a conserved sequence (consensus: EIRVGxxYQAxI; residues 166–177) that binds in a conserved groove on the HDAC. This is positioned close to the active site of the HDAC enzyme [26].
The carboxy-terminal region of the ELM2 domain forms a four-helix bundle with a small hydrophobic core and enlarges the interacting surface with HDAC1 as it completes a path around the ‘back’ of the enzyme. The four-helix bundle forms a homodimer and is therefore able to associate with a second HDAC1:MTA1 complex (Fig. 3). The homodimerisation interface is extensive with 14 non-polar side chains being contributed from each ELM2 domain. The highly complementary nature of this interface suggests that it is physiologically relevant. This means that the NuRD complex contains two copies of both HDAC1 and MTA1 proteins, and given that HDAC1 and HDAC2 are highly similar (83 % identical), it is likely that both HDACs can coexist in a single NuRD complex.
The dimerisation of HDAC1:MTA1 (through the ELM2 domain) positions the active site of the two HDACs on approximately the same face of the dimer. This orientation situates the two active sites approximately 90 Å apart and could allow the complex to simultaneously target more than one nucleosome.

4.3 The carboxy-terminus and interaction with RBBP4

RBBP4 and RBBP7 are WD40-repeat proteins that share 92 % identity and are integral components of the NuRD complex [2]. These proteins act as histone chaperones and associate with additional chromatin modifying complexes including SIN3A, PRC2 and NURF [4042]. RBBP4 and RBBP7 have a seven-bladed β-propeller architecture and have at least two distinct binding sites for partner proteins: the first on the top of the protein (structurally characterised as the binding site for histone H3 and FOG1) and the second on the side of the protein, involving α-helices at both the amino- and carboxy-termini and an extended loop inserted into blade 6 (the binding site for histone H4 and Su(z)12) [20, 43, 44].
Several studies have used GST pull-down assays to map the interaction between MTA1/2 and RBBP4/7. These have suggested that there may be three non-overlapping regions in the carboxy-terminus of MTA1 that mediate this interaction. One of these includes the GATA-type zinc finger domain [45, 46]. More recently, a short motif at the carboxy-terminus of MTA1 (residues 656–686) was structurally characterised as being able to bind to RBBP4 [25] (Fig. 4). This fragment is helical on binding to RBBP4 and binds in an acidic groove on the side of the WD40 domain. This groove was previously shown to bind to histone H4 [20]. Indeed, both fragments of MTA1 and histone H4 include the motif KRAARR and both form analogous contacts to RBBP4. Competition assays showed that MTA1 and histone H4 compete for the same binding site on RBBP4, suggesting that MTA1 modulates RBBP4 interaction with histones.
The second binding site was observed in the structure of the Drosophila homologue Nurf55 bound to a histone H3 tail peptide. In this structure, a peptide corresponding to histone H3 binds at the top of the propeller [43]. There is no overlap between the H3 and H4 binding sites, suggesting that MTA1 only competes for binding with histone H4. RBBP4 and RBBP7 are found in many corepressor complexes such as SIN3A, PRC2 and NURF, and the two distinct binding sites would increase their ability to recruit alternate binding partners in different settings.

5 Structural insight into other domains of the MTA family

MTA contains additional domains and motifs that remain to be structurally and functionally characterised. These domains have been identified through sequence alignment with other characterised proteins. Through sequence comparison and modelling, we can speculate on how these domains will look and how they will function.

5.1 The BAH domain

Hydrophobic cluster sequence analysis identified a single copy of the BAH domain in MTA1 that spans residues 1–164 [47]. The BAH domain was first described in the protein Polybromo, a subunit of a 2-MDa chromatin remodelling complex. Polybromo contains two BAH domains carboxy-terminal to four bromodomains [48]. The BAH domain has also been identified in a number of proteins involved in transcriptional regulation including the yeast RSC chromatin remodelling complex proteins (RSC1 and RSC2) and the transcription factor ASH1. BAH domains are also found in ORC1, SIR3 and DNMT1 proteins. Sequence alignments show that there are approximately 130 conserved amino acids within BAH domains although some contain large insertions of variable length.
Several structures of BAH domains have been solved and show that the core of the domain is largely β-sheet in character (Fig. 5). Beyond this core, each protein has a divergent fold including 310 helices, β-strands and ordered loops. This diversity may be a requirement of each protein to perform different functions. Structure prediction suggests that MTA1 will fold to form a β-barrel core with two long insertions.
The BAH domain of MTA1 is most closely related to the two BAH domains of Polybromo. The proximal BAH domain of Polybromo has a six-stranded β-barrel core, the centre of which is completely filled with hydrophobic residues [49]. A key feature of the domain is the long carboxy-terminal extension that effectively ‘wraps’ around the outside of the protein to stabilise two extended loops and the rest of the protein. Sequence comparison of the Polybromo-BAH domain with MTA1 suggests that the MTA1-BAH carboxy-terminal extension is shorter and is immediately followed by the ELM2 domain.
The BAH domains in RSC2, ORC1 and SIR3 have been shown to mediate interactions with chromatin. If this were also the case for the BAH domain from MTA1, this would provide an attractive explanation as to how chromatin is recruited to the corepressor complex allowing the HDAC to target acetylated lysines in the histone tails.
The BAH domain from the yeast RSC2 protein has been shown to bind histone H3 [50]. However, sequence comparison shows that the two tryptophan residues in the RSC2-BAH involved in binding the unmodified histone H3 peptide are not conserved in the MTA1-BAH domain suggesting that MTA1 cannot bind histone H3 in the same way as RSC2.
The yeast SIR3 protein not only interacts with histone tails but the BAH domain also makes extensive contacts with the nucleosome [5154]. The crystal structure of the complex shows that acetylated amino-terminus of SIR3-BAH domain is critical for interaction with histones H4 and H2B [53, 54]. The amino acids in SIR3 that mediate interactions with the nucleosome show very limited conservation in the MTA1-BAH domain, making it difficult to predict that MTA1 would bind nucleosomes in an analogous fashion.
The ORC1-BAH domain has also been shown to bind to histone H4. In this case, binding only occurs when the histone tail is dimethylated at lysine 20 (H4K20me2) [55]. The dimethylated side chain is bound within an aromatic cage on the surface of the ORC1-BAH domain. Again, sequence comparisons with the MTA1-BAH domain show that tyrosine and tryptophan residues important for binding H4K20me2 are not conserved.
These structures support the BAH domain as a histone-binding domain but the diversity within the family only allows a tentative suggestion that the BAH domain of MTA1 could bind to histone tails. Indeed, the two BAH domains from the chromatin-associated DNMT1 methyltransferase have not been shown to mediate direct interactions with chromatin [56]. Instead, they appear to contribute indirectly to substrate recognition by DNMT1.
Whilst it remains to be seen whether the MTA1-BAH domain directly recruits chromatin, the location of the domain immediately adjacent to the ELM2 domain means that it will be positioned very close to the active site of the HDAC enzyme [26]. Therefore, it seems inevitable that it will play some role in substrate interaction.

5.2 GATA-type zinc finger domain

MTA1 contains a GATA-type zinc finger domain positioned carboxy-terminal to the SANT domain. GATA-type zinc fingers are found in many eukaryotic transcription factors where they contribute to sequence-specific DNA recognition [57]. The zinc finger domain is composed of around 30 amino acids with four conserved cysteines coordinating a structural zinc ion [58] (Fig. 6). The domain interacts directly in the major groove of the DNA but also makes many interactions with the phosphate backbone of the DNA. The carboxy-terminus interacts in the adjacent minor groove of the DNA [58].
Sequence comparison shows that the GATA-type zinc finger in MTA1 is rather different from GATA1. It is likely that it retains DNA-binding activity, but given the lack of conservation of the relevant residues, it will probably either recognise a different sequence from GATA1 or, perhaps more likely, may bind to DNA without sequence specificity.

5.3 Additional domains and intrinsically disordered regions

MTA1 contains additional motifs that remain to be structurally and functionally characterised [31]. The xPPxP motif appears twice in MTA1 and could potentially be recognised by SH3 domains. The SH3 binding motif (xPPxP) is recognised by a 60 amino acid domain that is widely found in cytoskeletal elements and signalling proteins [59]. The proline-rich motif typically adopts a left-handed polyproline helix conformation and is recognised by the SH3 domain with characteristic β-barrel fold [60].
Towards the carboxy-terminus of MTA1, there are five SPXX motifs and these have been proposed to be sites of MTA1 phosphorylation [61]. The phosphorylation pattern on MTA1 was shown to dynamically change at each of these sites during the first 24 h of differentiation. One of the short forms of MTA1 (MTA1s) has been characterised as being phosphorylated by casein kinase [62]. The phosphorylation of MTA1 at this site was shown to be important for repressing estrogen-induced ER transactivation.
Further MTA1 motifs include a methylation site, which has shown to be involved in recruiting CHD4 and two nuclear localisation motifs [27, 63].

6 Conclusions

MTA1 is a key scaffold protein in the NuRD corepressor complex and plays a vital role in both assembling the complex and activating the HDAC enzymes. MTA1 directly contacts many of the core NuRD components. A number of these interactions occur within the structurally conserved domains of MTA1, but others are directed through intrinsically disordered regions that become ordered on binding and are key to forming a stable NuRD complex.
Three domains of MTA1 have now been characterised by NMR and X-ray crystallography. The ELM2 and SANT domains have been shown to recruit HDAC1 and/or HDAC2 through an extensive interface that excludes 1,278 Å2 from solvent exposure [26]. The extensive nature of this interface begins to address why the NuRD complex is so stable, as the components have previously been shown to remain intact on isolation from cells and show resistance to high salt treatment [3]. The stability of a core NuRD complex appears to be a common feature in other corepressor complexes such as the NCoR, SIN3 and PRC2 complexes [40, 41, 64].
The other structurally characterised motif (KRAARR) of MTA1 lies towards the carboxy-terminus. This motif has been shown to interact with RBBP4 and modulates binding to histone H4 [25]. This competition highlights the role of MTA1 as an essential scaffold component of the NuRD complex that mediates many key interactions.
In addition to its role as a scaffold protein, MTA1 also plays an important role in activating HDACs through forming one half of the binding site for inositol phosphates that have been shown to serve as activator molecules for class I HDACs.
Intriguingly, a many of the core NuRD complex proteins have the potential to interact with DNA and chromatin. The RBBP, p66, MTA and MBD proteins all have domains that are likely to interact with either histone tails, nucleosome or DNA. This permits the NuRD complex to use a large repertoire of modules to target nucleosome remodelling and deacetylase activity to chromatin. A full understanding of the activity and mechanism of the NuRD complex will require more challenging structural studies of the intact complex and how it interacts with its chromatin substrate. Such studies are likely also to provide further insights into strategies to target the complex therapeutically.

Acknowledgments

This work was supported by the Wellcome Trust (grant number WT085408) and the BBSRC (grant number 10867).
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Xue, Y., Wong, J., Moreno, G. T., Young, M. K., Côté, J., & Wang, W. (1998). NURD, a novel complex with both ATP-dependent chromatin-remodeling and histone deacetylase activities. Molecular Cell, 2(6), 851–861.PubMedCrossRef Xue, Y., Wong, J., Moreno, G. T., Young, M. K., Côté, J., & Wang, W. (1998). NURD, a novel complex with both ATP-dependent chromatin-remodeling and histone deacetylase activities. Molecular Cell, 2(6), 851–861.PubMedCrossRef
2.
Zurück zum Zitat Zhang, Y., Ng, H.-H., Erdjument-Bromage, H., Tempst, P., Bird, A., & Reinberg, D. (1999). Analysis of the NuRD subunits reveals a histone deacetylase core complex and a connection with DNA methylation. Genes and Development, 13(15), 1924–1935.PubMedCentralPubMedCrossRef Zhang, Y., Ng, H.-H., Erdjument-Bromage, H., Tempst, P., Bird, A., & Reinberg, D. (1999). Analysis of the NuRD subunits reveals a histone deacetylase core complex and a connection with DNA methylation. Genes and Development, 13(15), 1924–1935.PubMedCentralPubMedCrossRef
3.
Zurück zum Zitat Humphrey, G. W., Wang, Y., Russanova, V. R., Hirai, T., Qin, J., Nakatani, Y., & Howard, B. H. (2001). Stable histone deacetylase complexes distinguished by the presence of SANT domain proteins CoREST/kiaa0071 and Mta-L1. Journal of Biological Chemistry, 276(9), 6817–6824. doi:10.1074/jbc.M007372200.PubMedCrossRef Humphrey, G. W., Wang, Y., Russanova, V. R., Hirai, T., Qin, J., Nakatani, Y., & Howard, B. H. (2001). Stable histone deacetylase complexes distinguished by the presence of SANT domain proteins CoREST/kiaa0071 and Mta-L1. Journal of Biological Chemistry, 276(9), 6817–6824. doi:10.​1074/​jbc.​M007372200.PubMedCrossRef
4.
Zurück zum Zitat Wen, Y. D., Perissi, V., Staszewski, L. M., Yang, W. M., Krones, A., Glass, C. K., et al. (2000). The histone deacetylase-3 complex contains nuclear receptor corepressors. Proceedings of the National Academy of Sciences of the United States of America, 97(13), 7202–7207.PubMedCentralPubMedCrossRef Wen, Y. D., Perissi, V., Staszewski, L. M., Yang, W. M., Krones, A., Glass, C. K., et al. (2000). The histone deacetylase-3 complex contains nuclear receptor corepressors. Proceedings of the National Academy of Sciences of the United States of America, 97(13), 7202–7207.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Laherty, C. D., Yang, W. M., Sun, J. M., Davie, J. R., Seto, E., & Eisenman, R. N. (1997). Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression. Cell, 89(3), 349–356.PubMedCrossRef Laherty, C. D., Yang, W. M., Sun, J. M., Davie, J. R., Seto, E., & Eisenman, R. N. (1997). Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression. Cell, 89(3), 349–356.PubMedCrossRef
6.
Zurück zum Zitat Bantscheff, M., Hopf, C., Savitski, M. M., Dittmann, A., Grandi, P., Michon, A.-M., et al. (2011). Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nature Biotechnology, 29(3), 255–265. doi:10.1038/nbt.1759.PubMedCrossRef Bantscheff, M., Hopf, C., Savitski, M. M., Dittmann, A., Grandi, P., Michon, A.-M., et al. (2011). Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nature Biotechnology, 29(3), 255–265. doi:10.​1038/​nbt.​1759.PubMedCrossRef
7.
Zurück zum Zitat Guenther, M. G., Lane, W. S., Fischle, W., Verdin, E., Lazar, M. A., & Shiekhattar, R. (2000). A core SMRT corepressor complex containing HDAC3 and TBL1, a WD40-repeat protein linked to deafness. Genes and Development, 14(9), 1048–1057.PubMedCentralPubMed Guenther, M. G., Lane, W. S., Fischle, W., Verdin, E., Lazar, M. A., & Shiekhattar, R. (2000). A core SMRT corepressor complex containing HDAC3 and TBL1, a WD40-repeat protein linked to deafness. Genes and Development, 14(9), 1048–1057.PubMedCentralPubMed
11.
Zurück zum Zitat Toh, Y., Pencil, S. D., & Nicolson, G. L. (1994). A novel candidate metastasis-associated gene, mta1, differentially expressed in highly metastatic mammary adenocarcinoma cell lines. cDNA cloning, expression, and protein analyses. Journal of Biological Chemistry, 269(37), 22958–22963.PubMed Toh, Y., Pencil, S. D., & Nicolson, G. L. (1994). A novel candidate metastasis-associated gene, mta1, differentially expressed in highly metastatic mammary adenocarcinoma cell lines. cDNA cloning, expression, and protein analyses. Journal of Biological Chemistry, 269(37), 22958–22963.PubMed
12.
Zurück zum Zitat Toh, Y., & Nicolson, G. L. (2009). The role of the MTA family and their encoded proteins in human cancers: molecular functions and clinical implications. Clinical and Experimental Metastasis, 26(3), 215–227. doi:10.1007/s10585-008-9233-8.PubMedCrossRef Toh, Y., & Nicolson, G. L. (2009). The role of the MTA family and their encoded proteins in human cancers: molecular functions and clinical implications. Clinical and Experimental Metastasis, 26(3), 215–227. doi:10.​1007/​s10585-008-9233-8.PubMedCrossRef
14.
Zurück zum Zitat Zhang, H., Stephens, L. C., & Kumar, R. (2006). Metastasis tumor antigen family proteins during breast cancer progression and metastasis in a reliable mouse model for human breast cancer. Clinical Cancer Research an Official Journal of the American Association for Cancer Research, 12(5), 1479–1486. doi:10.1158/1078-0432.CCR-05-1519.PubMedCrossRef Zhang, H., Stephens, L. C., & Kumar, R. (2006). Metastasis tumor antigen family proteins during breast cancer progression and metastasis in a reliable mouse model for human breast cancer. Clinical Cancer Research an Official Journal of the American Association for Cancer Research, 12(5), 1479–1486. doi:10.​1158/​1078-0432.​CCR-05-1519.PubMedCrossRef
15.
18.
Zurück zum Zitat Wade, P. A., Jones, P. L., Vermaak, D., & Wolffe, A. P. (1998). A multiple subunit Mi-2 histone deacetylase from Xenopus laevis cofractionates with an associated Snf2 superfamily ATPase. Current Biology, 8(14), 843–846.PubMedCrossRef Wade, P. A., Jones, P. L., Vermaak, D., & Wolffe, A. P. (1998). A multiple subunit Mi-2 histone deacetylase from Xenopus laevis cofractionates with an associated Snf2 superfamily ATPase. Current Biology, 8(14), 843–846.PubMedCrossRef
23.
Zurück zum Zitat Zhang, Y., LeRoy, G., Seelig, H. P., Lane, W. S., & Reinberg, D. (1998). The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell, 95(2), 279–289.PubMedCrossRef Zhang, Y., LeRoy, G., Seelig, H. P., Lane, W. S., & Reinberg, D. (1998). The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell, 95(2), 279–289.PubMedCrossRef
24.
Zurück zum Zitat Toh, Y., Kuninaka, S., Endo, K., Oshiro, T., Ikeda, Y., Nakashima, H., et al. (2000). Molecular analysis of a candidate metastasis-associated gene, MTA1: possible interaction with histone deacetylase 1. Journal of Experimental and Clinical Cancer Research, 19(1), 105–111.PubMed Toh, Y., Kuninaka, S., Endo, K., Oshiro, T., Ikeda, Y., Nakashima, H., et al. (2000). Molecular analysis of a candidate metastasis-associated gene, MTA1: possible interaction with histone deacetylase 1. Journal of Experimental and Clinical Cancer Research, 19(1), 105–111.PubMed
25.
Zurück zum Zitat Alqarni, S. S. M., Murthy, A., Zhang, W., Przewloka, M. R., Silva, A. P. G., Watson, A. A., et al. (2014). Insight into the architecture of the NuRD complex: Structure of the RbAp48-MTA1 sub-complex. Journal of Biological Chemistry, 289(32), 21844–21855. doi:10.1074/jbc.M114.558940.PubMedCentralPubMedCrossRef Alqarni, S. S. M., Murthy, A., Zhang, W., Przewloka, M. R., Silva, A. P. G., Watson, A. A., et al. (2014). Insight into the architecture of the NuRD complex: Structure of the RbAp48-MTA1 sub-complex. Journal of Biological Chemistry, 289(32), 21844–21855. doi:10.​1074/​jbc.​M114.​558940.PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Yaguchi, M., Wada, Y., Toh, Y., Iguchi, H., Kono, A., Matsusue, K., & Takiguchi, S. (2005). Identification and characterization of the variants of metastasis-associated protein 1 generated following alternative splicing. Biochimica et Biophysica Acta, 1732(1–3), 8–14. doi:10.1016/j.bbaexp.2005.12.001.PubMedCrossRef Yaguchi, M., Wada, Y., Toh, Y., Iguchi, H., Kono, A., Matsusue, K., & Takiguchi, S. (2005). Identification and characterization of the variants of metastasis-associated protein 1 generated following alternative splicing. Biochimica et Biophysica Acta, 1732(1–3), 8–14. doi:10.​1016/​j.​bbaexp.​2005.​12.​001.PubMedCrossRef
29.
Zurück zum Zitat Kumar, R., Wang, R.-A., Mazumdar, A., Talukder, A. H., Mandal, M., Yang, Z., et al. (2002). A naturally occurring MTA1 variant sequesters oestrogen receptor-alpha in the cytoplasm. Nature, 418(6898), 654–657. doi:10.1038/nature00889.PubMedCrossRef Kumar, R., Wang, R.-A., Mazumdar, A., Talukder, A. H., Mandal, M., Yang, Z., et al. (2002). A naturally occurring MTA1 variant sequesters oestrogen receptor-alpha in the cytoplasm. Nature, 418(6898), 654–657. doi:10.​1038/​nature00889.PubMedCrossRef
30.
Zurück zum Zitat Kleene, R., Zdzieblo, J., Wege, K., & Kern, H. F. (1999). A novel zymogen granule protein (ZG29p) and the nuclear protein MTA1p are differentially expressed by alternative transcription initiation in pancreatic acinar cells of the rat. Journal of Cell Science, 112(Pt 15), 2539–2548.PubMed Kleene, R., Zdzieblo, J., Wege, K., & Kern, H. F. (1999). A novel zymogen granule protein (ZG29p) and the nuclear protein MTA1p are differentially expressed by alternative transcription initiation in pancreatic acinar cells of the rat. Journal of Cell Science, 112(Pt 15), 2539–2548.PubMed
31.
Zurück zum Zitat Nawa, A., Nishimori, K., Lin, P., Maki, Y., Moue, K., Sawada, H., et al. (2000). Tumor metastasis-associated human MTA1 gene: its deduced protein sequence, localization, and association with breast cancer cell proliferation using antisense phosphorothioate oligonucleotides. Journal of Cellular Biochemistry, 79(2), 202–212.PubMedCrossRef Nawa, A., Nishimori, K., Lin, P., Maki, Y., Moue, K., Sawada, H., et al. (2000). Tumor metastasis-associated human MTA1 gene: its deduced protein sequence, localization, and association with breast cancer cell proliferation using antisense phosphorothioate oligonucleotides. Journal of Cellular Biochemistry, 79(2), 202–212.PubMedCrossRef
32.
Zurück zum Zitat Solari, F., Bateman, A., & Ahringer, J. (1999). The Caenorhabditis elegans genes egl-27 and egr-1 are similar to MTA1, a member of a chromatin regulatory complex, and are redundantly required for embryonic patterning. Development, 126(11), 2483–2494.PubMed Solari, F., Bateman, A., & Ahringer, J. (1999). The Caenorhabditis elegans genes egl-27 and egr-1 are similar to MTA1, a member of a chromatin regulatory complex, and are redundantly required for embryonic patterning. Development, 126(11), 2483–2494.PubMed
33.
Zurück zum Zitat Nicolson, G. L., Nawa, A., Toh, Y., Taniguchi, S., Nishimori, K., & Moustafa, A. (2003). Tumor metastasis-associated human MTA1 gene and its MTA1 protein product: role in epithelial cancer cell invasion, proliferation and nuclear regulation. Clinical and Experimental Metastasis, 20(1), 19–24.PubMedCrossRef Nicolson, G. L., Nawa, A., Toh, Y., Taniguchi, S., Nishimori, K., & Moustafa, A. (2003). Tumor metastasis-associated human MTA1 gene and its MTA1 protein product: role in epithelial cancer cell invasion, proliferation and nuclear regulation. Clinical and Experimental Metastasis, 20(1), 19–24.PubMedCrossRef
34.
Zurück zum Zitat Boyer, L. A., Latek, R. R., & Peterson, C. L. (2004). The SANT domain: a unique histone-tail-binding module? Nature Reviews Molecular Cell Biology, 5(2), 158–163. doi:10.1038/nrm1314.PubMedCrossRef Boyer, L. A., Latek, R. R., & Peterson, C. L. (2004). The SANT domain: a unique histone-tail-binding module? Nature Reviews Molecular Cell Biology, 5(2), 158–163. doi:10.​1038/​nrm1314.PubMedCrossRef
35.
Zurück zum Zitat Aasland, R., Stewart, A. F., & Gibson, T. (1996). The SANT domain: a putative DNA-binding domain in the SWI-SNF and ADA complexes, the transcriptional co-repressor N-CoR and TFIIIB. Trends in Biochemical Sciences, 21(3), 87–88.PubMed Aasland, R., Stewart, A. F., & Gibson, T. (1996). The SANT domain: a putative DNA-binding domain in the SWI-SNF and ADA complexes, the transcriptional co-repressor N-CoR and TFIIIB. Trends in Biochemical Sciences, 21(3), 87–88.PubMed
36.
Zurück zum Zitat Ogata, K., Morikawa, S., Nakamura, H., Sekikawa, A., Inoue, T., Kanai, H., et al. (1994). Solution structure of a specific DNA complex of the Myb DNA-binding domain with cooperative recognition helices. Cell, 79(4), 639–648.PubMedCrossRef Ogata, K., Morikawa, S., Nakamura, H., Sekikawa, A., Inoue, T., Kanai, H., et al. (1994). Solution structure of a specific DNA complex of the Myb DNA-binding domain with cooperative recognition helices. Cell, 79(4), 639–648.PubMedCrossRef
37.
Zurück zum Zitat Guenther, M. G., Barak, O., & Lazar, M. A. (2001). The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Molecular and Cellular Biology, 21(18), 6091–6101.PubMedCentralPubMedCrossRef Guenther, M. G., Barak, O., & Lazar, M. A. (2001). The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Molecular and Cellular Biology, 21(18), 6091–6101.PubMedCentralPubMedCrossRef
39.
40.
Zurück zum Zitat Zhang, Y., Sun, Z. W., Iratni, R., Erdjument-Bromage, H., Tempst, P., Hampsey, M., & Reinberg, D. (1998). SAP30, a novel protein conserved between human and yeast, is a component of a histone deacetylase complex. Molecular Cell, 1(7), 1021–1031.PubMedCrossRef Zhang, Y., Sun, Z. W., Iratni, R., Erdjument-Bromage, H., Tempst, P., Hampsey, M., & Reinberg, D. (1998). SAP30, a novel protein conserved between human and yeast, is a component of a histone deacetylase complex. Molecular Cell, 1(7), 1021–1031.PubMedCrossRef
41.
Zurück zum Zitat Kuzmichev, A., Nishioka, K., Erdjument-Bromage, H., Tempst, P., & Reinberg, D. (2002). Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes and Development, 16(22), 2893–2905. doi:10.1101/gad.1035902.PubMedCentralPubMedCrossRef Kuzmichev, A., Nishioka, K., Erdjument-Bromage, H., Tempst, P., & Reinberg, D. (2002). Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes and Development, 16(22), 2893–2905. doi:10.​1101/​gad.​1035902.PubMedCentralPubMedCrossRef
44.
Zurück zum Zitat Lejon, S., Thong, S. Y., Murthy, A., AlQarni, S., Murzina, N. V., Blobel, G. A., et al. (2011). Insights into association of the NuRD complex with FOG-1 from the crystal structure of an RbAp48 FOG-1 complex. Journal of Biological Chemistry, 286(2), 1196–1203. doi:10.1074/jbc.M110.195842.PubMedCentralPubMedCrossRef Lejon, S., Thong, S. Y., Murthy, A., AlQarni, S., Murzina, N. V., Blobel, G. A., et al. (2011). Insights into association of the NuRD complex with FOG-1 from the crystal structure of an RbAp48 FOG-1 complex. Journal of Biological Chemistry, 286(2), 1196–1203. doi:10.​1074/​jbc.​M110.​195842.PubMedCentralPubMedCrossRef
45.
Zurück zum Zitat Roche, A. E., Bassett, B. J., Samant, S. A., Hong, W., Blobel, G. A., & Svensson, E. C. (2008). The zinc finger and C-terminal domains of MTA proteins are required for FOG-2-mediated transcriptional repression via the NuRD complex. Journal of Molecular and Cellular Cardiology, 44(2), 352–360. doi:10.1016/j.yjmcc.2007.10.023.PubMedCentralPubMedCrossRef Roche, A. E., Bassett, B. J., Samant, S. A., Hong, W., Blobel, G. A., & Svensson, E. C. (2008). The zinc finger and C-terminal domains of MTA proteins are required for FOG-2-mediated transcriptional repression via the NuRD complex. Journal of Molecular and Cellular Cardiology, 44(2), 352–360. doi:10.​1016/​j.​yjmcc.​2007.​10.​023.PubMedCentralPubMedCrossRef
47.
Zurück zum Zitat Callebaut, I., Courvalin, J. C., & Mornon, J. P. (1999). The BAH (bromo-adjacent homology) domain: a link between DNA methylation, replication and transcriptional regulation. FEBS Letters, 446(1), 189–193.PubMedCrossRef Callebaut, I., Courvalin, J. C., & Mornon, J. P. (1999). The BAH (bromo-adjacent homology) domain: a link between DNA methylation, replication and transcriptional regulation. FEBS Letters, 446(1), 189–193.PubMedCrossRef
48.
Zurück zum Zitat Nicolas, R. H., & Goodwin, G. H. (1996). Molecular cloning of polybromo, a nuclear protein containing multiple domains including five bromodomains, a truncated HMG-box, and two repeats of a novel domain. Gene, 175(1–2), 233–240.PubMedCrossRef Nicolas, R. H., & Goodwin, G. H. (1996). Molecular cloning of polybromo, a nuclear protein containing multiple domains including five bromodomains, a truncated HMG-box, and two repeats of a novel domain. Gene, 175(1–2), 233–240.PubMedCrossRef
49.
Zurück zum Zitat Oliver, A. W., Jones, S. A., Roe, S. M., Matthews, S., Goodwin, G. H., & Pearl, L. H. (2005). Crystal structure of the proximal BAH domain of the polybromo protein. Biochemical Journal, 389(Pt 3), 657–664. doi:10.1042/BJ20050310.PubMedCentralPubMed Oliver, A. W., Jones, S. A., Roe, S. M., Matthews, S., Goodwin, G. H., & Pearl, L. H. (2005). Crystal structure of the proximal BAH domain of the polybromo protein. Biochemical Journal, 389(Pt 3), 657–664. doi:10.​1042/​BJ20050310.PubMedCentralPubMed
52.
Zurück zum Zitat Wang, F., Li, G., Altaf, M., Lu, C., Currie, M. A., Johnson, A., & Moazed, D. (2013). Heterochromatin protein Sir3 induces contacts between the amino terminus of histone H4 and nucleosomal DNA. Proceedings of the National Academy of Sciences of the United States of America, 110(21), 8495–8500. doi:10.1073/pnas.1300126110.PubMedCentralPubMedCrossRef Wang, F., Li, G., Altaf, M., Lu, C., Currie, M. A., Johnson, A., & Moazed, D. (2013). Heterochromatin protein Sir3 induces contacts between the amino terminus of histone H4 and nucleosomal DNA. Proceedings of the National Academy of Sciences of the United States of America, 110(21), 8495–8500. doi:10.​1073/​pnas.​1300126110.PubMedCentralPubMedCrossRef
53.
Zurück zum Zitat Arnaudo, N., Fernández, I. S., McLaughlin, S. H., Peak-Chew, S. Y., Rhodes, D., & Martino, F. (2013). The N-terminal acetylation of Sir3 stabilizes its binding to the nucleosome core particle. Nature Structural & Molecular Biology, 20(9), 1119–1121. doi:10.1038/nsmb.2641.CrossRef Arnaudo, N., Fernández, I. S., McLaughlin, S. H., Peak-Chew, S. Y., Rhodes, D., & Martino, F. (2013). The N-terminal acetylation of Sir3 stabilizes its binding to the nucleosome core particle. Nature Structural & Molecular Biology, 20(9), 1119–1121. doi:10.​1038/​nsmb.​2641.CrossRef
54.
Zurück zum Zitat Yang, D., Fang, Q., Wang, M., Ren, R., Wang, H., He, M., et al. (2013). Nα-acetylated Sir3 stabilizes the conformation of a nucleosome-binding loop in the BAH domain. Nature Structural and Molecular Biology, 20(9), 1116–1118. doi:10.1038/nsmb.2637.PubMedCrossRef Yang, D., Fang, Q., Wang, M., Ren, R., Wang, H., He, M., et al. (2013). Nα-acetylated Sir3 stabilizes the conformation of a nucleosome-binding loop in the BAH domain. Nature Structural and Molecular Biology, 20(9), 1116–1118. doi:10.​1038/​nsmb.​2637.PubMedCrossRef
55.
56.
58.
Zurück zum Zitat Omichinski, J. G., Clore, G. M., Schaad, O., Felsenfeld, G., Trainor, C., Appella, E., et al. (1993). NMR structure of a specific DNA complex of Zn-containing DNA binding domain of GATA-1. Science, 261(5120), 438–446.PubMedCrossRef Omichinski, J. G., Clore, G. M., Schaad, O., Felsenfeld, G., Trainor, C., Appella, E., et al. (1993). NMR structure of a specific DNA complex of Zn-containing DNA binding domain of GATA-1. Science, 261(5120), 438–446.PubMedCrossRef
59.
Zurück zum Zitat Ren, R., Mayer, B. J., Cicchetti, P., & Baltimore, D. (1993). Identification of a ten-amino acid proline-rich SH3 binding site. Science, 259(5098), 1157–1161.PubMedCrossRef Ren, R., Mayer, B. J., Cicchetti, P., & Baltimore, D. (1993). Identification of a ten-amino acid proline-rich SH3 binding site. Science, 259(5098), 1157–1161.PubMedCrossRef
60.
Zurück zum Zitat Yu, H., Chen, J. K., Feng, S., Dalgarno, D. C., Brauer, A. W., & Schreiber, S. L. (1994). Structural basis for the binding of proline-rich peptides to SH3 domains. Cell, 76(5), 933–945.PubMedCrossRef Yu, H., Chen, J. K., Feng, S., Dalgarno, D. C., Brauer, A. W., & Schreiber, S. L. (1994). Structural basis for the binding of proline-rich peptides to SH3 domains. Cell, 76(5), 933–945.PubMedCrossRef
61.
Zurück zum Zitat Rigbolt, K. T. G., Prokhorova, T. A., Akimov, V., Henningsen, J., Johansen, P. T., Kratchmarova, I., et al. (2011). System-wide temporal characterization of the proteome and phosphoproteome of human embryonic stem cell differentiation. Science Signaling, 4(164), rs3. doi:10.1126/scisignal.2001570.PubMedCrossRef Rigbolt, K. T. G., Prokhorova, T. A., Akimov, V., Henningsen, J., Johansen, P. T., Kratchmarova, I., et al. (2011). System-wide temporal characterization of the proteome and phosphoproteome of human embryonic stem cell differentiation. Science Signaling, 4(164), rs3. doi:10.​1126/​scisignal.​2001570.PubMedCrossRef
62.
Zurück zum Zitat Mishra, S. K., Yang, Z., Mazumdar, A., Talukder, A. H., Larose, L., & Kumar, R. (2004). Metastatic tumor antigen 1 short form (MTA1s) associates with casein kinase I-gamma2, an estrogen-responsive kinase. Oncogene, 23(25), 4422–4429. doi:10.1038/sj.onc.1207569.PubMedCrossRef Mishra, S. K., Yang, Z., Mazumdar, A., Talukder, A. H., Larose, L., & Kumar, R. (2004). Metastatic tumor antigen 1 short form (MTA1s) associates with casein kinase I-gamma2, an estrogen-responsive kinase. Oncogene, 23(25), 4422–4429. doi:10.​1038/​sj.​onc.​1207569.PubMedCrossRef
63.
Zurück zum Zitat Toh, Y., Pencil, S. D., & Nicolson, G. L. (1995). Analysis of the complete sequence of the novel metastasis-associated candidate gene, mta1, differentially expressed in mammary adenocarcinoma and breast cancer cell lines. Gene, 159(1), 97–104.PubMedCrossRef Toh, Y., Pencil, S. D., & Nicolson, G. L. (1995). Analysis of the complete sequence of the novel metastasis-associated candidate gene, mta1, differentially expressed in mammary adenocarcinoma and breast cancer cell lines. Gene, 159(1), 97–104.PubMedCrossRef
64.
Zurück zum Zitat Zhang, J., Kalkum, M., Chait, B. T., & Roeder, R. G. (2002). The N-CoR-HDAC3 nuclear receptor corepressor complex inhibits the JNK pathway through the integral subunit GPS2. Molecular Cell, 9(3), 611–623.PubMedCrossRef Zhang, J., Kalkum, M., Chait, B. T., & Roeder, R. G. (2002). The N-CoR-HDAC3 nuclear receptor corepressor complex inhibits the JNK pathway through the integral subunit GPS2. Molecular Cell, 9(3), 611–623.PubMedCrossRef
65.
Zurück zum Zitat McGuffin, L. J., Bryson, K., & Jones, D. T. (2000). The PSIPRED protein structure prediction server. Bioinformatics, 16(4), 404–405.PubMedCrossRef McGuffin, L. J., Bryson, K., & Jones, D. T. (2000). The PSIPRED protein structure prediction server. Bioinformatics, 16(4), 404–405.PubMedCrossRef
Metadaten
Titel
Towards an understanding of the structure and function of MTA1
verfasst von
Christopher J. Millard
Louise Fairall
John W. R. Schwabe
Publikationsdatum
01.12.2014
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 4/2014
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-014-9513-5

Weitere Artikel der Ausgabe 4/2014

Cancer and Metastasis Reviews 4/2014 Zur Ausgabe

EditorialNotes

Preface

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.