Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 433(18): 167114, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34161779

ABSTRACT

Chromodomain-Helicase DNA binding protein 7 (CHD7) is an ATP dependent chromatin remodeler involved in maintaining open chromatin structure. Mutations of CHD7 gene causes multiple developmental disorders, notably CHARGE syndrome. However, there is not much known about the molecular mechanism by which CHD7 remodels nucleosomes. Here, we performed biochemical and biophysical analysis on CHD7 chromatin remodeler and uncover that N-terminal to the Chromodomain (N-CRD) interacts with nucleosome and contains a high conserved arginine stretch, which is reminiscent of arginine anchor. Importantly, this region is required for efficient ATPase stimulation and nucleosome remodeling activity of CHD7. Furthermore, smFRET analysis shows the mutations in the N-CRD causes the defects in remodeling activity. Collectively, our results uncover the functional importance of a previously unidentified N-terminal region in CHD7 and implicate that the multiple domains in chromatin remodelers are involved in regulating their activities.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Mutation , Nucleosomes , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Arginine/chemistry , Arginine/genetics , DNA Helicases/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Protein Conformation , Sequence Homology
2.
Nat Commun ; 12(1): 3014, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021162

ABSTRACT

Members of the chromodomain-helicase-DNA binding (CHD) protein family are chromatin remodelers implicated in human pathologies, with CHD6 being one of its least studied members. We discovered a de novo CHD6 missense mutation in a patient clinically presenting the rare Hallermann-Streiff syndrome (HSS). We used genome editing to generate isogenic iPSC lines and model HSS in relevant cell types. By combining genomics with functional in vivo and in vitro assays, we show that CHD6 binds a cohort of autophagy and stress response genes across cell types. The HSS mutation affects CHD6 protein folding and impairs its ability to recruit co-remodelers in response to DNA damage or autophagy stimulation. This leads to accumulation of DNA damage burden and senescence-like phenotypes. We therefore uncovered a molecular mechanism explaining HSS onset via chromatin control of autophagic flux and genotoxic stress surveillance.


Subject(s)
Autophagy/physiology , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Autophagy/genetics , Chromatin , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/metabolism , Epigenomics , Gene Editing , Gene Expression , Hallermann's Syndrome/genetics , Humans , Mutation , Phenotype
3.
Proc Natl Acad Sci U S A ; 117(46): 28847-28858, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127760

ABSTRACT

CHD7 encodes an ATP-dependent chromatin remodeling factor. Mutation of this gene causes multiple developmental disorders, including CHARGE (Coloboma of the eye, Heart defects, Atresia of the choanae, Retardation of growth/development, Genital abnormalities, and Ear anomalies) syndrome, in which conotruncal anomalies are the most prevalent form of heart defects. How CHD7 regulates conotruncal development remains unclear. In this study, we establish that deletion of Chd7 in neural crest cells (NCCs) causes severe conotruncal defects and perinatal lethality, thus providing mouse genetic evidence demonstrating that CHD7 cell-autonomously regulates cardiac NCC development, thereby clarifying a long-standing controversy in the literature. Using transcriptomic analyses, we show that CHD7 fine-tunes the expression of a gene network that is critical for cardiac NCC development. To gain further molecular insights into gene regulation by CHD7, we performed a protein-protein interaction screen by incubating recombinant CHD7 on a protein array. We find that CHD7 directly interacts with several developmental disorder-mutated proteins including WDR5, a core component of H3K4 methyltransferase complexes. This direct interaction suggested that CHD7 may recruit histone-modifying enzymes to target loci independently of its remodeling functions. We therefore generated a mouse model that harbors an ATPase-deficient allele and demonstrates that mutant CHD7 retains the ability to recruit H3K4 methyltransferase activity to its targets. Thus, our data uncover that CHD7 regulates cardiovascular development through ATP-dependent and -independent activities, shedding light on the etiology of CHD7-related congenital disorders. Importantly, our data also imply that patients carrying a premature stop codon versus missense mutations will likely display different molecular alterations; these patients might therefore require personalized therapeutic interventions.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart/embryology , Adenosine Triphosphate/metabolism , Alleles , Animals , CHARGE Syndrome/genetics , Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , Disease Models, Animal , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , Mutation , Neural Crest/embryology , Neural Crest/metabolism , Organogenesis/physiology
4.
Epigenetics Chromatin ; 12(1): 52, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434570

ABSTRACT

BACKGROUND: Maintenance of genome integrity during DNA replication is crucial to the perpetuation of all organisms. In eukaryotes, the bypass of DNA lesions by the replication machinery prevents prolonged stalling of the replication fork, which could otherwise lead to greater damages such as gross chromosomal rearrangements. Bypassing DNA lesions and subsequent repair are accomplished by the activation of DNA damage tolerance pathways such as the template switching (TS) pathway. In yeast, the RAD5 (Radiation-sensitive 5) protein plays a crucial role in initiating the TS pathway by catalyzing the polyubiquitination of PCNA (Proliferation Cell Nuclear Antigen). Likewise, one of the mammalian RAD5-homologs, SHPRH (SNF2, histone linker, PHD, RING, helicase) mediates PCNA polyubiquitination. To date, the study of SHPRH enzymatic functions has been limited to this modification. It is therefore unclear how SHPRH carries out its function in DNA repair. Moreover, how this protein regulates gene transcription at the enzymatic level is also unknown. RESULTS: Given that SHPRH harbors domains found in chromatin remodeling proteins, we investigated its biochemical properties in the presence of nucleosomal substrates. We find that SHPRH binds equally well to double-stranded (ds) DNA and to nucleosome core particles, however, like ISWI and CHD-family remodelers, SHPRH shows a strong preference for nucleosomes presenting extranucleosomal DNA. Moreover, nucleosomes but not dsDNA strongly stimulate the ATPase activity of SHPRH. Intriguingly, unlike typically observed with SNF2-family enzymes, ATPase activity does not translate into conventional nucleosome remodeling, under standard assay conditions. To test whether SHPRH can act as a ubiquitin E3 ligase for nucleosomes, we performed a screen using 26 E2-conjugating enzymes. We uncover that SHPRH is a potent nucleosome E3-ubiquitin-ligase that can function with at least 7 different E2s. Mass spectrometry analyses of products generated in the presence of the UBE2D1-conjugating enzyme reveal that SHPRH can catalyze the formation of polyubiquitin linkages that are either branched or associated with the recruitment of DNA repair factors, as well as linkages involved in proteasomal degradation. CONCLUSIONS: We propose that, in addition to polyubiquitinating PCNA, SHPRH promotes DNA repair or transcriptional regulation in part through chromatin ubiquitination. Our study sets a biochemical framework for studying other RAD5- and RAD16-related protein functions through the ubiquitination of nucleosomes.


Subject(s)
DNA Helicases/metabolism , Nucleosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases/metabolism , DNA/metabolism , DNA Repair , Histones/metabolism , Humans , Proliferating Cell Nuclear Antigen/metabolism , Substrate Specificity , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
5.
Org Biomol Chem ; 17(7): 1827-1833, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30604825

ABSTRACT

We report a high-affinity photoswitchable DNA binder, which displays different nucleosome-binding capacities upon visible-light irradiation. Both photochemical and DNA-recognition properties were examined by UV-Vis, HPLC, CD spectroscopy, NMR, FID assays, EMSA and DLS. Our probe sets the basis for developing new optoepigenetic tools for conditional modulation of nucleosomal DNA accessibility.


Subject(s)
Azo Compounds/chemistry , DNA/chemistry , Intercalating Agents/chemistry , Light , Nucleosomes/chemistry , Azo Compounds/chemical synthesis , Intercalating Agents/chemical synthesis , Molecular Structure , Photochemical Processes
6.
Nat Commun ; 8: 14806, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28378812

ABSTRACT

Gene regulation by steroid hormones plays important roles in health and disease. In Drosophila, the hormone ecdysone governs transitions between key developmental stages. Ecdysone-regulated genes are bound by a heterodimer of ecdysone receptor (EcR) and Ultraspiracle. According to the bimodal switch model, steroid hormone receptors recruit corepressors in the absence of hormone and coactivators in its presence. Here we show that the nucleosome remodeller dMi-2 is recruited to ecdysone-regulated genes to limit transcription. Contrary to the prevalent model, recruitment of the dMi-2 corepressor increases upon hormone addition to constrain gene activation through chromatin remodelling. Furthermore, EcR and dMi-2 form a complex that is devoid of Ultraspiracle. Unexpectedly, EcR contacts the dMi-2 ATPase domain and increases the efficiency of dMi-2-mediated nucleosome remodelling. This study identifies a non-canonical EcR-corepressor complex with the potential for a direct regulation of ATP-dependent nucleosome remodelling by a nuclear hormone receptor.


Subject(s)
Adenosine Triphosphatases/physiology , Autoantigens/physiology , Drosophila Proteins/physiology , Ecdysone/physiology , Gene Expression Regulation/physiology , Receptors, Steroid/physiology , Transcription, Genetic/physiology , Adenosine Triphosphatases/metabolism , Animals , Chromatin/metabolism , Drosophila/genetics , Ecdysone/metabolism , Kinetics , Transcriptional Activation
7.
Mol Cell ; 51(4): 454-68, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23911928

ABSTRACT

DNA damage is linked to multiple human diseases, such as cancer, neurodegeneration, and aging. Little is known about the role of chromatin accessibility in DNA repair. Here, we find that the deacetylase sirtuin 6 (SIRT6) is one of the earliest factors recruited to double-strand breaks (DSBs). SIRT6 recruits the chromatin remodeler SNF2H to DSBs and focally deacetylates histone H3K56. Lack of SIRT6 and SNF2H impairs chromatin remodeling, increasing sensitivity to genotoxic damage and recruitment of downstream factors such as 53BP1 and breast cancer 1 (BRCA1). Remarkably, SIRT6-deficient mice exhibit lower levels of chromatin-associated SNF2H in specific tissues, a phenotype accompanied by DNA damage. We demonstrate that SIRT6 is critical for recruitment of a chromatin remodeler as an early step in the DNA damage response, indicating that proper unfolding of chromatin plays a rate-limiting role. We present a unique crosstalk between a histone modifier and a chromatin remodeler, regulating a coordinated response to prevent DNA damage.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/genetics , DNA Repair/genetics , Genomic Instability , Sirtuins/metabolism , Sirtuins/physiology , Adenosine Triphosphatases/genetics , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Hippocampus/cytology , Hippocampus/metabolism , Histones/metabolism , Humans , Immunoprecipitation , Mice , Mice, Knockout , Nucleosomes/metabolism , Sirtuins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
Elife ; 2: e01270, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23986864

ABSTRACT

Biochemical assays reveal that nucleosome maturation and chromatin remodelling by the motor protein Chd1 are distinct, separable enzymatic activities.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Animals , Humans
9.
Proc Natl Acad Sci U S A ; 109(47): 19238-43, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23134727

ABSTRACT

Mutations in the CHD7 gene cause human developmental disorders including CHARGE syndrome. Genetic studies in model organisms have further established CHD7 as a central regulator of vertebrate development. Functional analysis of the CHD7 protein has been hampered by its large size. We used a dual-tag system to purify intact recombinant CHD7 protein and found that it is an ATP-dependent nucleosome remodeling factor. Biochemical analyses indicate that CHD7 has characteristics distinct from SWI/SNF- and ISWI-type remodelers. Further investigations show that CHD7 patient mutations have consequences that range from subtle to complete inactivation of remodeling activity, and that mutations leading to protein truncations upstream of amino acid 1899 of CHD7 are likely to cause a hypomorphic phenotype for remodeling. We propose that nucleosome remodeling is a key function for CHD7 during developmental processes and provide a molecular basis for predicting the impact of disease mutations on that function.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Point Mutation/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acids/genetics , CHARGE Syndrome/genetics , Chromosomal Proteins, Non-Histone/metabolism , Humans , Hypogonadism/genetics , Mutant Proteins/metabolism , Nucleosomes/metabolism , Transcription Factors/metabolism
10.
Curr Protoc Mol Biol ; Chapter 21: Unit 21.17.1-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20069538

ABSTRACT

Methylation-sensitive single-molecule analysis of chromatin structure is a high-resolution method for studying nucleosome positioning. As described in this unit, this method allows for the analysis of the chromatin structure of unmethylated CpG islands or in vitro-remodeled nucleosomes by treatment with the CpG-specific DNA methyltransferase SssI (M.SssI), followed by bisulfite sequencing of individual progeny DNA molecules. Unlike nuclease-based approaches, this method allows each molecule to be viewed as an individual entity instead of an average population.


Subject(s)
Chromatin/chemistry , DNA Methylation , Genetic Techniques , Animals , Biocatalysis , CpG Islands , Humans
11.
Nucleic Acids Res ; 37(16): 5279-94, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19567737

ABSTRACT

Chromatin remodeling enzymes use the energy of ATP hydrolysis to alter histone-DNA contacts and regulate DNA-based processes in eukaryotes. Whether different subfamilies of remodeling complexes generate distinct products remains uncertain. We have developed a protocol to analyze nucleosome remodeling on individual products formed in vitro. We used a DNA methyltransferase to examine DNA accessibility throughout nucleosomes that had been remodeled by the ISWI and SWI/SNF families of enzymes. We confirmed that ISWI-family enzymes mainly created patterns of accessibility consistent with canonical nucleosomes. In contrast, SWI/SNF-family enzymes generated widespread DNA accessibility. The protection patterns created by these enzymes were usually located at the extreme ends of the DNA and showed no evidence for stable loop formation on individual molecules. Instead, SWI/SNF family proteins created extensive accessibility by generating heterogeneous products that had fewer histone-DNA contacts than a canonical nucleosome, consistent with models in which a canonical histone octamer has been 'pushed' off of the end of the DNA.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Nucleosomes/chemistry , Adenosine Triphosphate/metabolism , DNA/chemistry , DNA Helicases/metabolism , DNA-Cytosine Methylases , Histones/chemistry , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Transcription Factors/metabolism
12.
Chromosome Res ; 14(4): 433-49, 2006.
Article in English | MEDLINE | ID: mdl-16821138

ABSTRACT

The regulation of chromatin structure is of fundamental importance for many DNA-based processes in eukaryotes. Activation or repression of gene transcription or DNA replication depends on enzymes which can generate the appropriate chromatin environment. Several of these enzymes utilize the energy of ATP hydrolysis to alter nucleosome structure. In recent years our understanding of the multisubunit complexes within which they function, their mechanisms of action, their regulation and their in-vivo roles has increased. Much of what we have learned has been gleaned from studies in Drosophila melanogaster. Here we will review what we know about the main classes of ATP-dependent chromatin remodelers in Drosophila.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/physiology , Animals , Autoantigens , Chromatin Assembly and Disassembly/physiology , DNA Helicases , Dyneins , Homeodomain Proteins
13.
J Biol Chem ; 280(51): 41912-20, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16223721

ABSTRACT

A plethora of ATP-dependent chromatin-remodeling enzymes have been identified during the last decade. Many have been shown to play pivotal roles in the organization and expression of eukaryotic genomes. It is clear that their activities need to be tightly regulated to ensure their coordinated action. However, little is known about how ATP-dependent remodelers are regulated at the molecular level. Here, we have investigated the ATP-dependent chromatin remodeling enzyme Mi-2 of Drosophila melanogaster. Radioactive labeling of S2 cells reveals that dMi-2 is a phosphoprotein in vivo. dMi-2 phosphorylation is constitutive, and we identify dCK2 as a major dMi-2 kinase in cell extracts. dCK2 binds to and phosphorylates a dMi-2 N-terminal region. Dephosphorylation of recombinant dMi-2 increases its affinity for the nucleosome substrate, nucleosome-stimulated ATPase, and ATP-dependent nucleosome mobilization activities. Our results reveal a potential mechanism for regulation of the dMi-2 enzyme and point toward CK2 phosphorylation as a common feature of CHD family ATPases.


Subject(s)
Adenosine Triphosphatases/metabolism , Autoantigens/metabolism , Chromatin/metabolism , Cyclin-Dependent Kinase 2/metabolism , Drosophila Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Cell Line , Chromatography, Affinity , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/isolation & purification , Drosophila melanogaster , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Phosphorylation , Protein Binding , Sequence Homology, Amino Acid
15.
EMBO J ; 21(10): 2430-40, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12006495

ABSTRACT

Drosophila Mi-2 (dMi-2) is the ATPase subunit of a complex combining ATP-dependent nucleosome remodelling and histone deacetylase activities. dMi-2 contains an HMG box-like region, two PHD fingers, two chromodomains and a SNF2-type ATPase domain. It is not known which of these domains contribute to nucleosome remodelling. We have tested a panel of dMi-2 deletion mutants in ATPase, nucleosome mobilization and nucleosome binding assays. Deletion of the chromodomains impairs all three activities. A dMi-2 mutant lacking the chromodomains is incorporated into a functional histone deacetylase complex in vivo but has lost nucleosome-stimulated ATPase activity. In contrast to dHP1, dMi-2 does not bind methylated histone H3 tails and does not require histone tails for nucleosome binding. Instead, the dMi-2 chromodomains display DNA binding activity that is not shared by other chromodomains. Our results suggest that the chromodomains act at an early step of the remodelling process to bind the nucleosome substrate predominantly via protein-DNA interactions. Furthermore, we identify DNA binding as a novel chromodomain-associated activity.


Subject(s)
Adenosine Triphosphate/metabolism , Autoantigens/chemistry , Autoantigens/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila/metabolism , Nucleosomes/physiology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , DNA-Binding Proteins/chemistry , Histones/analysis , Molecular Sequence Data , Protein Subunits , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...