Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Nucleic Acids Res ; 52(17): 10194-10219, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39142653

ABSTRACT

The chromatin-remodeling enzyme helicase lymphoid-specific (HELLS) interacts with cell division cycle-associated 7 (CDCA7) on nucleosomes and is involved in the regulation of DNA methylation in higher organisms. Mutations in these genes cause immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome, which also results in DNA hypomethylation of satellite repeat regions. We investigated the functional domains of human CDCA7 in HELLS using several mutant CDCA7 proteins. The central region is critical for binding to HELLS, activation of ATPase, and nucleosome sliding activities of HELLS-CDCA7. The N-terminal region tends to inhibit ATPase activity. The C-terminal 4CXXC-type zinc finger domain contributes to CpG and hemimethylated CpG DNA preference for DNA-dependent HELLS-CDCA7 ATPase activity. Furthermore, CDCA7 showed a binding preference to DNA containing hemimethylated CpG, and replication-dependent pericentromeric heterochromatin foci formation of CDCA7 with HELLS was observed in mouse embryonic stem cells; however, all these phenotypes were lost in the case of an ICF syndrome mutant of CDCA7 mutated in the zinc finger domain. Thus, CDCA7 most likely plays a role in the recruitment of HELLS, activates its chromatin remodeling function, and efficiently induces DNA methylation, especially at hemimethylated replication sites.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases , DNA Methylation , Zinc Fingers , Humans , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/chemistry , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/metabolism , CpG Islands/genetics , DNA/metabolism , DNA/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Mutation , Protein Binding , Nucleosomes/metabolism , Nucleosomes/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Protein Domains , Mouse Embryonic Stem Cells/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Heterochromatin/metabolism , Heterochromatin/genetics , Face/abnormalities , Nuclear Proteins
2.
Mol Cell ; 66(5): 711-720.e3, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28529057

ABSTRACT

The multidomain CCCTC-binding factor (CTCF), containing a tandem array of 11 zinc fingers (ZFs), modulates the three-dimensional organization of chromatin. We crystallized the human CTCF DNA-binding domain in complex with a known CTCF-binding site. While ZF2 does not make sequence-specific contacts, each finger of ZF3-7 contacts three bases of the 15-bp consensus sequence. Each conserved nucleotide makes base-specific hydrogen bonds with a particular residue. Most of the variable base pairs within the core sequence also engage in interactions with the protein. These interactions compensate for deviations from the consensus sequence, allowing CTCF to adapt to sequence variations. CTCF is sensitive to cytosine methylation at position 2, but insensitive at position 12 of the 15-bp core sequence. These differences can be rationalized structurally. Although included in crystallizations, ZF10 and ZF11 are not visible, while ZF8 and ZF9 span the backbone of the DNA duplex, conferring no sequence specificity but adding to overall binding stability.


Subject(s)
DNA Methylation , DNA/metabolism , Repressor Proteins/metabolism , 5-Methylcytosine/metabolism , Binding Sites , CCCTC-Binding Factor , Cloning, Molecular , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , Humans , Hydrogen Bonding , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Stability , Repressor Proteins/chemistry , Repressor Proteins/genetics , Structure-Activity Relationship , Trinucleotide Repeats , Zinc Fingers
3.
Genes Dev ; 28(20): 2304-13, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25258363

ABSTRACT

In mammalian DNA, cytosine occurs in several chemical forms, including unmodified cytosine (C), 5-methylcytosine (5 mC), 5-hydroxymethylcytosine (5 hmC), 5-formylcytosine (5 fC), and 5-carboxylcytosine (5 caC). 5 mC is a major epigenetic signal that acts to regulate gene expression. 5 hmC, 5 fC, and 5 caC are oxidized derivatives that might also act as distinct epigenetic signals. We investigated the response of the zinc finger DNA-binding domains of transcription factors early growth response protein 1 (Egr1) and Wilms tumor protein 1 (WT1) to different forms of modified cytosine within their recognition sequence, 5'-GCG(T/G)GGGCG-3'. Both displayed high affinity for the sequence when C or 5 mC was present and much reduced affinity when 5 hmC or 5 fC was present, indicating that they differentiate primarily oxidized C from unoxidized C, rather than methylated C from unmethylated C. 5 caC affected the two proteins differently, abolishing binding by Egr1 but not by WT1. We ascribe this difference to electrostatic interactions in the binding sites. In Egr1, a negatively charged glutamate conflicts with the negatively charged carboxylate of 5 caC, whereas the corresponding glutamine of WT1 interacts with this group favorably. Our analyses shows that zinc finger proteins (and their splice variants) can respond in modulated ways to alternative modifications within their binding sequence.


Subject(s)
Cytosine/analogs & derivatives , WT1 Proteins/chemistry , WT1 Proteins/genetics , WT1 Proteins/metabolism , Crystallization , Cytosine/metabolism , DNA Methylation , Early Growth Response Protein 1/chemistry , Early Growth Response Protein 1/metabolism , Humans , Models, Molecular , Mutation , Oxidation-Reduction , Protein Binding , Protein Structure, Tertiary
4.
Nucleic Acids Res ; 47(16): 8388-8398, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31226215

ABSTRACT

ZBTB24, encoding a protein of the ZBTB family of transcriptional regulators, is one of four known genes-the other three being DNMT3B, CDCA7 and HELLS-that are mutated in immunodeficiency, centromeric instability and facial anomalies (ICF) syndrome, a genetic disorder characterized by DNA hypomethylation and antibody deficiency. The molecular mechanisms by which ZBTB24 regulates gene expression and the biological functions of ZBTB24 are poorly understood. Here, we identified a 12-bp consensus sequence [CT(G/T)CCAGGACCT] occupied by ZBTB24 in the mouse genome. The sequence is present at multiple loci, including the Cdca7 promoter region, and ZBTB24 binding is mostly associated with gene activation. Crystallography and DNA-binding data revealed that the last four of the eight zinc fingers (ZFs) (i.e. ZF5-8) in ZBTB24 confer specificity of DNA binding. Two ICF missense mutations have been identified in the ZBTB24 ZF domain, which alter zinc-binding cysteine residues. We demonstrated that the corresponding C382Y and C407G mutations in mouse ZBTB24 abolish specific DNA binding and fail to induce Cdca7 expression. Our analyses indicate and suggest a structural basis for the sequence specific recognition by a transcription factor centrally important for the pathogenesis of ICF syndrome.


Subject(s)
Cell Cycle Proteins/genetics , Face/abnormalities , Genome , Mutation, Missense , Nuclear Proteins/genetics , Primary Immunodeficiency Diseases/genetics , Repressor Proteins/chemistry , Transcription Factors/chemistry , Zinc Fingers/genetics , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Face/pathology , Gene Expression , Genetic Loci , Genetic Vectors , Humans , Mice , Models, Molecular , Nuclear Proteins/metabolism , Nucleotide Motifs , Primary Immunodeficiency Diseases/metabolism , Primary Immunodeficiency Diseases/pathology , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nature ; 506(7488): 391-5, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24390346

ABSTRACT

Cytosine residues in mammalian DNA occur in five forms: cytosine (C), 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). The ten-eleven translocation (Tet) dioxygenases convert 5mC to 5hmC, 5fC and 5caC in three consecutive, Fe(II)- and α-ketoglutarate-dependent oxidation reactions. The Tet family of dioxygenases is widely distributed across the tree of life, including in the heterolobosean amoeboflagellate Naegleria gruberi. The genome of Naegleria encodes homologues of mammalian DNA methyltransferase and Tet proteins. Here we study biochemically and structurally one of the Naegleria Tet-like proteins (NgTet1), which shares significant sequence conservation (approximately 14% identity or 39% similarity) with mammalian Tet1. Like mammalian Tet proteins, NgTet1 acts on 5mC and generates 5hmC, 5fC and 5caC. The crystal structure of NgTet1 in complex with DNA containing a 5mCpG site revealed that NgTet1 uses a base-flipping mechanism to access 5mC. The DNA is contacted from the minor groove and bent towards the major groove. The flipped 5mC is positioned in the active-site pocket with planar stacking contacts, Watson-Crick polar hydrogen bonds and van der Waals interactions specific for 5mC. The sequence conservation between NgTet1 and mammalian Tet1, including residues involved in structural integrity and functional significance, suggests structural conservation across phyla.


Subject(s)
5-Methylcytosine/metabolism , DNA/chemistry , DNA/metabolism , Dioxygenases/chemistry , Dioxygenases/metabolism , Naegleria/enzymology , 5-Methylcytosine/chemistry , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Conserved Sequence , Crystallography, X-Ray , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/chemistry , HEK293 Cells , Humans , Hydrogen Bonding , Mice , Mixed Function Oxygenases/chemistry , Models, Molecular , Molecular Sequence Data , Naegleria/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
6.
Nucleic Acids Res ; 45(5): 2396-2407, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27903915

ABSTRACT

The oncogenic transcription factor MYC and its binding partner MAX regulate gene expression by binding to DNA at enhancer-box (E-box) elements 5΄-CACGTG-3΄. In mammalian genomes, the central E-box CpG has the potential to be methylated at the 5-position of cytosine (5mC), or to undergo further oxidation to the 5-hydroxymethyl (5hmC), 5-formyl (5fC), or 5-carboxyl (5caC) forms. We find that MAX exhibits the greatest affinity for a 5caC or unmodified C-containing E-box, and much reduced affinities for the corresponding 5mC, 5hmC or 5fC forms. Crystallization of MAX with a 5caC modified E-box oligonucleotide revealed that MAX Arg36 recognizes 5caC using a 5caC-Arg-Guanine triad, with the next nearest residue to the carboxylate group being Arg60. In an analysis of >800 primary multiple myelomas, MAX alterations occurred at a frequency of ∼3%, more than half of which were single nucleotide substitutions affecting a basic clamp-like interface important for DNA interaction. Among these, arginines 35, 36 and 60 were the most frequently altered. In vitro binding studies showed that whereas mutation of Arg36 (R36W) or Arg35 (R35H/L) completely abolished DNA binding, mutation of Arg60 (R60Q) significantly reduced DNA binding, but retained a preference for the 5caC modified E-box. Interestingly, MAX alterations define a subset of myeloma patients with lower MYC expression and a better overall prognosis. Together these data indicate that MAX can act as a direct epigenetic sensor of E-box cytosine modification states and that local CpG modification and MAX variants converge to modulate the MAX-MYC transcriptional network.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cytosine/analogs & derivatives , E-Box Elements , Multiple Myeloma/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Arginine/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CpG Islands , Cytosine/chemistry , Cytosine/metabolism , DNA/chemistry , DNA/metabolism , Epigenesis, Genetic , Mutation , Protein Binding , Protein Conformation , Repressor Proteins/metabolism
7.
Nucleic Acids Res ; 44(21): 10165-10176, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27596598

ABSTRACT

Mutations in human zinc-finger transcription factor WT1 result in abnormal development of the kidneys and genitalia and an array of pediatric problems including nephropathy, blastoma, gonadal dysgenesis and genital discordance. Several overlapping phenotypes are associated with WT1 mutations, including Wilms tumors, Denys-Drash syndrome (DDS), Frasier syndrome (FS) and WAGR syndrome (Wilms tumor, aniridia, genitourinary malformations, and mental retardation). These conditions vary in severity from individual to individual; they can be fatal in early childhood, or relatively benign into adulthood. DDS mutations cluster predominantly in zinc fingers (ZF) 2 and 3 at the C-terminus of WT1, which together with ZF4 determine the sequence-specificity of DNA binding. We examined three DDS associated mutations in ZF2 of human WT1 where the normal glutamine at position 369 is replaced by arginine (Q369R), lysine (Q369K) or histidine (Q369H). These mutations alter the sequence-specificity of ZF2, we find, changing its affinity for certain bases and certain epigenetic forms of cytosine. X-ray crystallography of the DNA binding domains of normal WT1, Q369R and Q369H in complex with preferred sequences revealed the molecular interactions responsible for these affinity changes. DDS is inherited in an autosomal dominant fashion, implying a gain of function by mutant WT1 proteins. This gain, we speculate, might derive from the ability of the mutant proteins to sequester WT1 into unproductive oligomers, or to erroneously bind to variant target sequences.


Subject(s)
Denys-Drash Syndrome/genetics , Epigenesis, Genetic , Mutation , WT1 Proteins/genetics , WT1 Proteins/metabolism , Adenine/metabolism , Amino Acid Substitution , Crystallography, X-Ray , Cytosine/chemistry , Cytosine/metabolism , DNA/chemistry , DNA/metabolism , Glutamine/genetics , Guanine/metabolism , Humans , WT1 Proteins/chemistry
8.
Nucleic Acids Res ; 44(21): 10177-10185, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27596594

ABSTRACT

Reprogramming of mammalian genome methylation is critically important but poorly understood. Klf4, a transcription factor directing reprogramming, contains a DNA binding domain with three consecutive C2H2 zinc fingers. Klf4 recognizes CpG or TpG within a specific sequence. Mouse Klf4 DNA binding domain has roughly equal affinity for methylated CpG or TpG, and slightly lower affinity for unmodified CpG. The structural basis for this key preference is unclear, though the side chain of Glu446 is known to contact the methyl group of 5-methylcytosine (5mC) or thymine (5-methyluracil). We examined the role of Glu446 by mutagenesis. Substituting Glu446 with aspartate (E446D) resulted in preference for unmodified cytosine, due to decreased affinity for 5mC. In contrast, substituting Glu446 with proline (E446P) increased affinity for 5mC by two orders of magnitude. Structural analysis revealed hydrophobic interaction between the proline's aliphatic cyclic structure and the 5-methyl group of the pyrimidine (5mC or T). As in wild-type Klf4 (E446), the proline at position 446 does not interact directly with either the 5mC N4 nitrogen or the thymine O4 oxygen. In contrast, the unmethylated cytosine's exocyclic N4 amino group (NH2) and its ring carbon C5 atom hydrogen bond directly with the aspartate carboxylate of the E446D variant. Both of these interactions would provide a preference for cytosine over thymine, and the latter one could explain the E446D preference for unmethylated cytosine. Finally, we evaluated the ability of these Klf4 mutants to regulate transcription of methylated and unmethylated promoters in a luciferase reporter assay.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mutation , 5-Methylcytosine/metabolism , Animals , CpG Islands , Crystallography, X-Ray , DNA/metabolism , DNA Methylation , Fluorescence Polarization , HEK293 Cells , Humans , Hydrogen Bonding , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/chemistry , Mice , Proline/metabolism , Protein Conformation , Thymine/metabolism
9.
Proc Natl Acad Sci U S A ; 112(4): 949-56, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25561520

ABSTRACT

Fragile X syndrome (FXS) results in intellectual disability (ID) most often caused by silencing of the fragile X mental retardation 1 (FMR1) gene. The resulting absence of fragile X mental retardation protein 1 (FMRP) leads to both pre- and postsynaptic defects, yet whether the pre- and postsynaptic functions of FMRP are independent and have distinct roles in FXS neuropathology remain poorly understood. Here, we demonstrate an independent presynaptic function for FMRP through the study of an ID patient with an FMR1 missense mutation. This mutation, c.413G > A (R138Q), preserves FMRP's canonical functions in RNA binding and translational regulation, which are traditionally associated with postsynaptic compartments. However, neuronally driven expression of the mutant FMRP is unable to rescue structural defects at the neuromuscular junction in fragile x mental retardation 1 (dfmr1)-deficient Drosophila, suggesting a presynaptic-specific impairment. Furthermore, mutant FMRP loses the ability to rescue presynaptic action potential (AP) broadening in Fmr1 KO mice. The R138Q mutation also disrupts FMRP's interaction with the large-conductance calcium-activated potassium (BK) channels that modulate AP width. These results reveal a presynaptic- and translation-independent function of FMRP that is linked to a specific subset of FXS phenotypes.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome , Mutation, Missense , Seizures , Action Potentials/genetics , Amino Acid Substitution , Animals , Child , Child, Preschool , Drosophila , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Gene Expression Regulation/genetics , Humans , Male , Mice , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Seizures/physiopathology
10.
Hum Mol Genet ; 24(6): 1733-40, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25416280

ABSTRACT

Fragile X syndrome, a common cause of intellectual disability and autism, is due to mutational silencing of the FMR1 gene leading to the absence of its gene product, fragile X mental retardation protein (FMRP). FMRP is a selective RNA binding protein owing to two central K-homology domains and a C-terminal arginine-glycine-glycine (RGG) box. However, several properties of the FMRP amino terminus are unresolved. It has been documented for over a decade that the amino terminus has the ability to bind RNA despite having no recognizable functional motifs. Moreover, the amino terminus has recently been shown to bind chromatin and influence the DNA damage response as well as function in the presynaptic space, modulating action potential duration. We report here the amino terminal crystal structures of wild-type FMRP, and a mutant (R138Q) that disrupts the amino terminus function, containing an integral tandem Agenet and discover a novel KH motif.


Subject(s)
Fragile X Mental Retardation Protein/chemistry , Amino Acid Motifs , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Humans , Mutation, Missense , Protein Structure, Tertiary
11.
Nucleic Acids Res ; 43(22): 10713-21, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26323320

ABSTRACT

The family of ten-eleven translocation (Tet) dioxygenases is widely distributed across the eukaryotic tree of life, from mammals to the amoeboflagellate Naegleria gruberi. Like mammalian Tet proteins, the Naegleria Tet-like protein, NgTet1, acts on 5-methylcytosine (5mC) and generates 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) in three consecutive, Fe(II)- and α-ketoglutarate-dependent oxidation reactions. The two intermediates, 5hmC and 5fC, could be considered either as the reaction product of the previous enzymatic cycle or the substrate for the next cycle. Here we present a new crystal structure of NgTet1 in complex with DNA containing a 5hmC. Along with the previously solved NgTet1-5mC structure, the two complexes offer a detailed picture of the active site at individual stages of the reaction cycle. In the crystal, the hydroxymethyl (OH-CH2-) moiety of 5hmC points to the metal center, representing the reaction product of 5mC hydroxylation. The hydroxyl oxygen atom could be rotated away from the metal center, to a hydrophobic pocket formed by Ala212, Val293 and Phe295. Such rotation turns the hydroxyl oxygen atom away from the product conformation, and exposes the target CH2 towards the metal-ligand water molecule, where a dioxygen O2 molecule would occupy to initiate the next round of reaction by abstracting a hydrogen atom from the substrate. The Ala212-to-Val (A212V) mutant profoundly limits the product to 5hmC, probably because the reduced hydrophobic pocket size restricts the binding of 5hmC as a substrate.


Subject(s)
Cytosine/analogs & derivatives , DNA/chemistry , Dioxygenases/chemistry , Naegleria/enzymology , 5-Methylcytosine/chemistry , Amino Acid Substitution , Catalytic Domain , Cytosine/chemistry , Dioxygenases/genetics , Models, Molecular , Thymine/chemistry
12.
J Biol Chem ; 290(34): 20723-20733, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26152719

ABSTRACT

One of the most fundamental questions in the control of gene expression in mammals is how the patterns of epigenetic modifications of DNA are generated, recognized, and erased. This includes covalent cytosine methylation of DNA and its associated oxidation states. An array of AdoMet-dependent methyltransferases, Fe(II)- and α-ketoglutarate-dependent dioxygenases, base excision glycosylases, and sequence-specific transcription factors is responsible for changing, maintaining, and interpreting the modification status of specific regions of chromatin. This review focuses on recent developments in characterizing the functional and structural links between the modification status of two DNA bases 5-methylcytosine and thymine (5-methyluracil).


Subject(s)
5-Methylcytosine/metabolism , DNA Glycosylases/metabolism , Dioxygenases/metabolism , Methyltransferases/metabolism , Protein Processing, Post-Translational , Thymine/metabolism , Transcription Factors/metabolism , DNA Glycosylases/genetics , DNA Methylation , DNA Repair , Dioxygenases/chemistry , Dioxygenases/genetics , Epigenesis, Genetic , Gene Expression , Humans , Iron/metabolism , Ketoglutaric Acids/metabolism , Methyltransferases/genetics , Models, Molecular , Oxidation-Reduction , Transcription Factors/genetics
13.
Nucleic Acids Res ; 42(8): 4859-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24520114

ABSTRACT

Transcription factor Krüppel-like factor 4 (Klf4), one of the factors directing cellular reprogramming, recognizes the CpG dinucleotide (whether methylated or unmodified) within a specific G/C-rich sequence. The binding affinity of the mouse Klf4 DNA-binding domain for methylated DNA is only slightly stronger than that for an unmodified oligonucleotide. The structure of the C-terminal three Krüppel-like zinc fingers (ZnFs) of mouse Klf4, in complex with fully methylated DNA, was determined at 1.85 Å resolution. An arginine and a glutamate interact with the methyl group. By comparison with two other recently characterized structures of ZnF protein complexes with methylated DNA, we propose a common principle of recognition of methylated CpG by C2H2 ZnF proteins, which involves a spatially conserved Arg-Glu pair.


Subject(s)
DNA Methylation , Kruppel-Like Transcription Factors/chemistry , Amino Acid Sequence , Animals , Arginine/chemistry , CpG Islands , Glutamic Acid/chemistry , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Molecular Sequence Data , Protein Binding , Sequence Alignment , Transcription Factors/chemistry
14.
Nature ; 455(7214): 826-9, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18772888

ABSTRACT

Maintenance methylation of hemimethylated CpG dinucleotides at DNA replication forks is the key to faithful mitotic inheritance of genomic methylation patterns. UHRF1 (ubiquitin-like, containing PHD and RING finger domains 1) is required for maintenance methylation by interacting with DNA nucleotide methyltransferase 1 (DNMT1), the maintenance methyltransferase, and with hemimethylated CpG, the substrate for DNMT1 (refs 1 and 2). Here we present the crystal structure of the SET and RING-associated (SRA) domain of mouse UHRF1 in complex with DNA containing a hemimethylated CpG site. The DNA is contacted in both the major and minor grooves by two loops that penetrate into the middle of the DNA helix. The 5-methylcytosine has flipped completely out of the DNA helix and is positioned in a binding pocket with planar stacking contacts, Watson-Crick polar hydrogen bonds and van der Waals interactions specific for 5-methylcytosine. Hence, UHRF1 contains a previously unknown DNA-binding module and is the first example of a non-enzymatic, sequence-specific DNA-binding protein domain to use the base flipping mechanism to interact with DNA.


Subject(s)
5-Methylcytosine/metabolism , DNA Methylation , DNA/chemistry , DNA/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , CpG Islands/genetics , Crystallography, X-Ray , DNA/genetics , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Conformation , Protein Binding , Protein Structure, Tertiary , Ubiquitin-Protein Ligases
15.
Nucleic Acids Res ; 40(17): 8276-84, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22740654

ABSTRACT

The mammalian DNA glycosylase--methyl-CpG binding domain protein 4 (MBD4)--is involved in active DNA demethylation via the base excision repair pathway. MBD4 contains an N-terminal MBD and a C-terminal DNA glycosylase domain. MBD4 can excise the mismatched base paired with a guanine (G:X), where X is uracil, thymine or 5-hydroxymethyluracil (5hmU). These are, respectively, the deamination products of cytosine, 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC). Here, we present three structures of the MBD4 C-terminal glycosylase domain (wild-type and its catalytic mutant D534N), in complex with DNA containing a G:T or G:5hmU mismatch. MBD4 flips the target nucleotide from the double-stranded DNA. The catalytic mutant D534N captures the intact target nucleotide in the active site binding pocket. MBD4 specifically recognizes the Watson-Crick polar edge of thymine or 5hmU via the O2, N3 and O4 atoms, thus restricting its activity to thymine/uracil-based modifications while excluding cytosine and its derivatives. The wild-type enzyme cleaves the N-glycosidic bond, leaving the ribose ring in the flipped state, while the cleaved base is released. Unexpectedly, the C1' of the sugar has yet to be hydrolyzed and appears to form a stable intermediate with one of the side chain carboxyl oxygen atoms of D534, via either electrostatic or covalent interaction, suggesting a different catalytic mechanism from those of other DNA glycosylases.


Subject(s)
Base Pair Mismatch , DNA Glycosylases/chemistry , Endodeoxyribonucleases/chemistry , Pentoxyl/analogs & derivatives , Thymine/chemistry , Animals , DNA/chemistry , DNA/metabolism , Endodeoxyribonucleases/metabolism , Mice , Models, Molecular , Pentoxyl/chemistry , Pentoxyl/metabolism , Protein Structure, Tertiary , Thymine/metabolism
16.
Nucleic Acids Res ; 40(20): 10203-14, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22962365

ABSTRACT

The mammalian thymine DNA glycosylase (TDG) is implicated in active DNA demethylation via the base excision repair pathway. TDG excises the mismatched base from G:X mismatches, where X is uracil, thymine or 5-hydroxymethyluracil (5hmU). These are, respectively, the deamination products of cytosine, 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC). In addition, TDG excises the Tet protein products 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) but not 5hmC and 5mC, when paired with a guanine. Here we present a post-reactive complex structure of the human TDG domain with a 28-base pair DNA containing a G:5hmU mismatch. TDG flips the target nucleotide from the double-stranded DNA, cleaves the N-glycosidic bond and leaves the C1' hydrolyzed abasic sugar in the flipped state. The cleaved 5hmU base remains in a binding pocket of the enzyme. TDG allows hydrogen-bonding interactions to both T/U-based (5hmU) and C-based (5caC) modifications, thus enabling its activity on a wider range of substrates. We further show that the TDG catalytic domain has higher activity for 5caC at a lower pH (5.5) as compared to the activities at higher pH (7.5 and 8.0) and that the structurally related Escherichia coli mismatch uracil glycosylase can excise 5caC as well. We discuss several possible mechanisms, including the amino-imino tautomerization of the substrate base that may explain how TDG discriminates against 5hmC and 5mC.


Subject(s)
Cytosine/analogs & derivatives , DNA/chemistry , Pentoxyl/analogs & derivatives , Thymine DNA Glycosylase/chemistry , Biocatalysis , Catalytic Domain , Cytosine/chemistry , Cytosine/metabolism , DNA/metabolism , Humans , Models, Molecular , Pentoxyl/chemistry , Pentoxyl/metabolism , Thymine DNA Glycosylase/metabolism
17.
Nucleic Acids Res ; 40(11): 4841-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362737

ABSTRACT

Cytosine residues in mammalian DNA occur in at least three forms, cytosine (C), 5-methylcytosine (M; 5mC) and 5-hydroxymethylcytosine (H; 5hmC). During semi-conservative DNA replication, hemi-methylated (M/C) and hemi-hydroxymethylated (H/C) CpG dinucleotides are transiently generated, where only the parental strand is modified and the daughter strand contains native cytosine. Here, we explore the role of DNA methyltransferases (DNMT) and ten eleven translocation (Tet) proteins in perpetuating these states after replication, and the molecular basis of their recognition by methyl-CpG-binding domain (MBD) proteins. Using recombinant proteins and modified double-stranded deoxyoligonucleotides, we show that DNMT1 prefers a hemi-methylated (M/C) substrate (by a factor of >60) over hemi-hydroxymethylated (H/C) and unmodified (C/C) sites, whereas both DNMT3A and DNMT3B have approximately equal activity on all three substrates (C/C, M/C and H/C). Binding of MBD proteins to methylated DNA inhibited Tet1 activity, suggesting that MBD binding may also play a role in regulating the levels of 5hmC. All five MBD proteins generally have reduced binding affinity for 5hmC relative to 5mC in the fully modified context (H/M versus M/M), though their relative abilities to distinguish the two varied considerably. We further show that the deamination product of 5hmC could be excised by thymine DNA glycosylase and MBD4 glycosylases regardless of context.


Subject(s)
Cytosine/analogs & derivatives , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Replication , 5-Methylcytosine/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferase 1 , DNA-Binding Proteins/metabolism , Humans , Pentoxyl/analogs & derivatives , Pentoxyl/metabolism , Thymine DNA Glycosylase/metabolism
18.
Nat Commun ; 15(1): 2467, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503750

ABSTRACT

In higher eukaryotes, a single DOT1 histone H3 lysine 79 (H3K79) methyltransferase processively produces H3K79me2/me3 through histone H2B mono-ubiquitin interaction, while the kinetoplastid Trypanosoma brucei di-methyltransferase DOT1A and tri-methyltransferase DOT1B efficiently methylate the homologous H3K76 without H2B mono-ubiquitination. Based on structural and biochemical analyses of DOT1A, we identify key residues in the methyltransferase motifs VI and X for efficient ubiquitin-independent H3K76 methylation in kinetoplastids. Substitution of a basic to an acidic residue within motif VI (Gx6K) is essential to stabilize the DOT1A enzyme-substrate complex, while substitution of the motif X sequence VYGE by CAKS renders a rigid active-site loop flexible, implying a distinct mechanism of substrate recognition. We further reveal distinct methylation kinetics and substrate preferences of DOT1A (H3K76me0) and DOT1B (DOT1A products H3K76me1/me2) in vitro, determined by a Ser and Ala residue within motif IV, respectively, enabling DOT1A and DOT1B to mediate efficient H3K76 tri-methylation non-processively but cooperatively, and suggesting why kinetoplastids have evolved two DOT1 enzymes.


Subject(s)
Histones , Ubiquitin , Histones/metabolism , Lysine/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methylation
19.
Nucleic Acids Res ; 38(11): 3533-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20156997

ABSTRACT

In eukaryotic nuclei, DNA is wrapped around an octamer of core histones to form nucleosomes, and chromatin fibers are thought to be stabilized by linker histones of the H1 type. Higher eukaryotes express multiple variants of histone H1; chickens possess six H1 variants. Here, we generated and analyzed the phenotype of a complete deletion of histone H1 genes in chicken cells. The H1-null cells showed decreased global nucleosome spacing, expanded nuclear volumes, and increased chromosome aberration rates, although proper mitotic chromatin structure appeared to be maintained. Expression array analysis revealed that the transcription of multiple genes was affected and was mostly downregulated in histone H1-deficient cells. This report describes the first histone H1 complete knockout cells in vertebrates and suggests that linker histone H1, while not required for mitotic chromatin condensation, plays important roles in nucleosome spacing and interphase chromatin compaction and acts as a global transcription regulator.


Subject(s)
Histones/physiology , Nucleosomes/chemistry , Animals , Cell Cycle , Cell Line , Chickens/genetics , Chickens/growth & development , Chickens/metabolism , Chromatin/ultrastructure , Chromosome Aberrations , Histones/genetics , Interphase/genetics , Mutation , Transcription, Genetic
20.
Sci Rep ; 12(1): 17691, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271106

ABSTRACT

In Saccharomyces cerevisiae, the pre-mRNA leakage 39-kDa protein (ScPml39) was reported to retain unspliced pre-mRNA prior to export through nuclear pore complexes (NPCs). Pml39 homologs outside the Saccharomycetaceae family are currently unknown, and mechanistic insight into Pml39 function is lacking. Here we determined the crystal structure of ScPml39 at 2.5 Å resolution to facilitate the discovery of orthologs beyond Saccharomycetaceae, e.g. in Schizosaccharomyces pombe or human. The crystal structure revealed integrated zf-C3HC and Rsm1 modules, which are tightly associated through a hydrophobic interface to form a single domain. Both zf-C3HC and Rsm1 modules belong to the Zn-containing BIR (Baculovirus IAP repeat)-like super family, with key residues of the canonical BIR domain being conserved. Features unique to the Pml39 modules refer to the spacing between the Zn-coordinating residues, giving rise to a substantially tilted helix αC in the zf-C3HC and Rsm1 modules, and an extra helix αAB' in the Rsm1 module. Conservation of key residues responsible for its distinct features identifies S. pombe Rsm1 and Homo sapiens NIPA/ZC3HC1 as structural orthologs of ScPml39. Based on the recent functional characterization of NIPA/ZC3HC1 as a scaffold protein that stabilizes the nuclear basket of the NPC, our data suggest an analogous function of ScPml39 in S. cerevisiae.


Subject(s)
Nuclear Proteins , Saccharomyces cerevisiae Proteins , Humans , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/chemistry , RNA Precursors/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL