Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
J Biol Chem ; 298(2): 101558, 2022 02.
Article in English | MEDLINE | ID: mdl-34979096

ABSTRACT

Chromatin-modifying complexes containing histone deacetylase (HDAC) activities play critical roles in the regulation of gene transcription in eukaryotes. These complexes are thought to lack intrinsic DNA-binding activity, but according to a well-established paradigm, they are recruited via protein-protein interactions by gene-specific transcription factors and posttranslational histone modifications to their sites of action on the genome. The mammalian Sin3L/Rpd3L complex, comprising more than a dozen different polypeptides, is an ancient HDAC complex found in diverse eukaryotes. The subunits of this complex harbor conserved domains and motifs of unknown structure and function. Here, we show that Sds3, a constitutively-associated subunit critical for the proper functioning of the Sin3L/Rpd3L complex, harbors a type of Tudor domain that we designate the capped Tudor domain. Unlike canonical Tudor domains that bind modified histones, the Sds3 capped Tudor domain binds to nucleic acids that can form higher-order structures such as G-quadruplexes and shares similarities with the knotted Tudor domain of the Esa1 histone acetyltransferase that was previously shown to bind single-stranded RNA. Our findings expand the range of macromolecules capable of recruiting the Sin3L/Rpd3L complex and draw attention to potentially new biological roles for this HDAC complex.


Subject(s)
G-Quadruplexes , Histone Deacetylases , Sin3 Histone Deacetylase and Corepressor Complex , Amino Acid Sequence , Animals , Histone Deacetylases/metabolism , Mammals , Protein Binding , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Transcription Factors/metabolism , Tudor Domain
2.
FEBS Lett ; 594(14): 2322-2330, 2020 07.
Article in English | MEDLINE | ID: mdl-32391601

ABSTRACT

The Sin3L/Rpd3L histone deacetylase (HDAC) complex is one of six major HDAC complexes in the nucleus, and its recruitment by promoter-bound transcription factors is an important step in many gene transcription regulatory pathways. Here, we investigate how the Myt1L zinc finger transcription factor, important for neuronal differentiation and the maintenance of neuronal identity, recruits this complex at the molecular level. We show that Myt1L, through a highly conserved segment shared with its paralogs, interacts directly and specifically with the Sin3 PAH1 domain, binding principally to the canonical hydrophobic cleft found in paired amphipathic helix domain (PAH) domains. Our findings are relevant not only for other members of the Myt family but also for enhancing our understanding of the rules of protein-protein interactions involving Sin3 PAH domains.


Subject(s)
Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Nerve Tissue Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/chemistry , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Humans , Mice , Models, Molecular , Protein Binding , Protein Domains
3.
J Biol Chem ; 294(38): 13928-13938, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31358618

ABSTRACT

The constitutively nuclear histone deacetylases (HDACs) 1, 2, and 3 erase acetyl marks on acetyllysine residues, alter the landscape of histone modifications, and modulate chromatin structure and dynamics and thereby crucially regulate gene transcription in higher eukaryotes. Nuclear HDACs exist as at least six giant multiprotein complexes whose nonenzymatic subunits confer genome targeting specificity for these enzymes. The deacetylase activity of HDACs has been shown previously to be enhanced by inositol phosphates, which also bridge the catalytic domain in protein-protein interactions with SANT (Swi3, Ada2, N-Cor, and TFIIIB) domains in all HDAC complexes except those that contain the Sin3 transcriptional corepressors. Here, using purified recombinant proteins, coimmunoprecipitation and HDAC assays, and pulldown and NMR experiments, we show that HDAC1/2 deacetylase activity in one of the most ancient and evolutionarily conserved Sin3L/Rpd3L complexes is inducibly up-regulated by inositol phosphates but involves interactions with a zinc finger motif in the Sin3-associated protein 30 (SAP30) subunit that is structurally unrelated to SANT domains, indicating convergent evolution at the functional level. This implies that this mode of regulation has evolved independently multiple times and provides an evolutionary advantage. We also found that constitutive association with another core subunit, Rb-binding protein 4 chromatin-binding factor (RBBP4), further enhances deacetylase activity, implying both inducible and constitutive regulatory mechanisms within the same HDAC complex. Our results indicate that inositol phosphates stimulate HDAC activity and that the SAP30 zinc finger motif performs roles similar to that of the unrelated SANT domain in promoting the SAP30-HDAC1 interaction and enhancing HDAC activity.


Subject(s)
Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Inositol Phosphates/metabolism , Amino Acid Sequence , DNA-Binding Proteins/metabolism , HEK293 Cells , Histone Deacetylase 1/ultrastructure , Histone Deacetylases/ultrastructure , Humans , Inositol Phosphates/chemistry , Inositol Phosphates/physiology , Models, Theoretical , Nuclear Receptor Co-Repressor 1 , Protein Binding , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/metabolism , Transcription Factors , Transcriptional Activation , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL