Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Biophys Rev ; 16(3): 365-382, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39099839

ABSTRACT

Pioneer transcription factors are proteins with a dual function. First, they regulate transcription by binding to nucleosome-free DNA regulatory elements. Second, they bind to DNA while wrapped around histone proteins in the chromatin and mediate chromatin opening. The molecular mechanisms that connect the two functions are yet to be discovered. In recent years, pioneer factors received increased attention mainly because of their crucial role in promoting cell fate transitions that could be used for regenerative therapies. For example, the three factors required to induce pluripotency in somatic cells, Oct4, Sox2, and Klf4 were classified as pioneer factors and studied extensively. With this increased attention, several structures of complexes between pioneer factors and chromatin structural units (nucleosomes) have been resolved experimentally. Furthermore, experimental and computational approaches have been designed to study two unresolved, key scientific questions: First, do pioneer factors induce directly local opening of nucleosomes and chromatin fibers upon binding? And second, how do the unstructured tails of the histones impact the structural dynamics involved in such conformational transitions? Here we review the current knowledge about transcription factor-induced nucleosome dynamics and the role of the histone tails in this process. We discuss what is needed to bridge the gap between the static views obtained from the experimental structures and the key structural dynamic events in chromatin opening. Finally, we propose that integrating nuclear magnetic resonance spectroscopy with molecular dynamics simulations is a powerful approach to studying pioneer factor-mediated dynamics of nucleosomes and perhaps small chromatin fibers using native DNA sequences.

2.
Cell Stem Cell ; 31(1): 127-147.e9, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38141611

ABSTRACT

Our understanding of pluripotency remains limited: iPSC generation has only been established for a few model species, pluripotent stem cell lines exhibit inconsistent developmental potential, and germline transmission has only been demonstrated for mice and rats. By swapping structural elements between Sox2 and Sox17, we built a chimeric super-SOX factor, Sox2-17, that enhanced iPSC generation in five tested species: mouse, human, cynomolgus monkey, cow, and pig. A swap of alanine to valine at the interface between Sox2 and Oct4 delivered a gain of function by stabilizing Sox2/Oct4 dimerization on DNA, enabling generation of high-quality OSKM iPSCs capable of supporting the development of healthy all-iPSC mice. Sox2/Oct4 dimerization emerged as the core driver of naive pluripotency with its levels diminished upon priming. Transient overexpression of the SK cocktail (Sox+Klf4) restored the dimerization and boosted the developmental potential of pluripotent stem cells across species, providing a universal method for naive reset in mammals.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Mice , Rats , Animals , Swine , Macaca fascicularis/metabolism , Induced Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Cellular Reprogramming , SOXB1 Transcription Factors/metabolism , Cell Differentiation , Mammals/metabolism
3.
Nat Commun ; 14(1): 787, 2023 02 11.
Article in English | MEDLINE | ID: mdl-36774438

ABSTRACT

During influenza A virus (IAV) infections, viral proteins are targeted by cellular E3 ligases for modification with ubiquitin. Here, we decipher and functionally explore the ubiquitination landscape of the IAV polymerase proteins during infection of human alveolar epithelial cells by applying mass spectrometry analysis of immuno-purified K-ε-GG (di-glycyl)-remnant-bearing peptides. We have identified 59 modified lysines across the three subunits, PB2, PB1 and PA of the viral polymerase of which 17 distinctively affect mRNA transcription, vRNA replication and the generation of recombinant viruses via non-proteolytic mechanisms. Moreover, further functional and in silico analysis indicate that ubiquitination at K578 in the PB1 thumb domain is mechanistically linked to dynamic structural transitions of the viral polymerase that are required for vRNA replication. Mutations K578A and K578R differentially affect the generation of recombinant viruses by impeding cRNA and vRNA synthesis, NP binding as well as polymerase dimerization. Collectively, our results demonstrate that the ubiquitin-mediated charge neutralization at PB1-K578 disrupts the interaction to an unstructured loop in the PB2 N-terminus that is required to coordinate polymerase dimerization and facilitate vRNA replication. This provides evidence that IAV exploits the cellular ubiquitin system to modulate the activity of the viral polymerase for viral replication.


Subject(s)
Influenza A virus , Influenza, Human , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Viral Proteins/metabolism , Transcription, Genetic , Nucleotidyltransferases/metabolism , Virus Replication , Ubiquitination , Ubiquitins/metabolism , RNA, Viral/genetics
4.
Nucleic Acids Res ; 51(3): 1120-1138, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36631980

ABSTRACT

Oct4 is essential to maintain pluripotency and has a pivotal role in establishing the germline. Its DNA-binding POU domain was recently found to bind motifs with methylated CpG elements normally associated with epigenetic silencing. However, the mode of binding and the consequences of this capability has remained unclear. Here, we show that Oct4 binds to a compact palindromic DNA element with a methylated CpG core (CpGpal) in alternative states of pluripotency and during cellular reprogramming towards induced pluripotent stem cells (iPSCs). During cellular reprogramming, typical Oct4 bound enhancers are uniformly demethylated, with the prominent exception of the CpGpal sites where DNA methylation is often maintained. We demonstrate that Oct4 cooperatively binds the CpGpal element as a homodimer, which contrasts with the ectoderm-expressed POU factor Brn2. Indeed, binding to CpGpal is Oct4-specific as other POU factors expressed in somatic cells avoid this element. Binding assays combined with structural analyses and molecular dynamic simulations show that dimeric Oct4-binding to CpGpal is driven by the POU-homeodomain whilst the POU-specific domain is detached from DNA. Collectively, we report that Oct4 exerts parts of its regulatory function in the context of methylated DNA through a DNA recognition mechanism that solely relies on its homeodomain.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Octamer Transcription Factor-3 , Cell Differentiation/genetics , DNA/metabolism , DNA Methylation , Epigenesis, Genetic , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Humans , Animals , Mice
5.
Nucleic Acids Res ; 50(18): 10311-10327, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36130732

ABSTRACT

Pioneer transcription factors are proteins that induce cellular identity transitions by binding to inaccessible regions of DNA in nuclear chromatin. They contribute to chromatin opening and recruit other factors to regulatory DNA elements. The structural features and dynamics modulating their interaction with nucleosomes are still unresolved. From a combination of experiments and molecular simulations, we reveal here how the pioneer factor and master regulator of pluripotency, Oct4, interprets and enhances nucleosome structural flexibility. The magnitude of Oct4's impact on nucleosome dynamics depends on the binding site position and the mobility of the unstructured tails of nucleosomal histone proteins. Oct4 uses both its DNA binding domains to propagate and stabilize open nucleosome conformations, one for specific sequence recognition and the other for nonspecific interactions with nearby regions of DNA. Our findings provide a structural basis for the versatility of transcription factors in engaging with nucleosomes and have implications for understanding how pioneer factors induce chromatin dynamics.


Subject(s)
Nucleosomes , Octamer Transcription Factor-3/metabolism , Chromatin/genetics , Histones/metabolism , Nucleosomes/genetics , Transcription Factors/metabolism
6.
PLoS Comput Biol ; 17(6): e1009013, 2021 06.
Article in English | MEDLINE | ID: mdl-34081696

ABSTRACT

Genomic DNA is packaged in chromatin, a dynamic fiber variable in size and compaction. In chromatin, repeating nucleosome units wrap 145-147 DNA basepairs around histone proteins. Genetic and epigenetic regulation of genes relies on structural transitions in chromatin which are driven by intra- and inter-nucleosome dynamics and modulated by chemical modifications of the unstructured terminal tails of histones. Here we demonstrate how the interplay between histone H3 and H2A tails control ample nucleosome breathing motions. We monitored large openings of two genomic nucleosomes, and only moderate breathing of an engineered nucleosome in atomistic molecular simulations amounting to 24 µs. Transitions between open and closed nucleosome conformations were mediated by the displacement and changes in compaction of the two histone tails. These motions involved changes in the DNA interaction profiles of clusters of epigenetic regulatory aminoacids in the tails. Removing the histone tails resulted in a large increase of the amplitude of nucleosome breathing but did not change the sequence dependent pattern of the motions. Histone tail modulated nucleosome breathing is a key mechanism of chromatin dynamics with important implications for epigenetic regulation.


Subject(s)
Genomics , Histones/metabolism , Nucleosomes/metabolism , Cluster Analysis , DNA/metabolism , Epigenesis, Genetic , Molecular Dynamics Simulation , Nucleic Acid Conformation
7.
J Mol Biol ; 433(6): 166744, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33309853

ABSTRACT

Gene regulation programs establish cellular identity and rely on dynamic changes in the structural packaging of genomic DNA. The DNA is packaged in chromatin, which is formed from arrays of nucleosomes displaying different degree of compaction and different lengths of inter-nucleosomal linker DNA. The nucleosome represents the repetitive unit of chromatin and is formed by wrapping 145-147 basepairs of DNA around an octamer of histone proteins. Each of the four histones is present twice and has a structured core and intrinsically disordered terminal tails. Chromatin dynamics are triggered by inter- and intra-nucleosome motions that are controlled by the DNA sequence, the interactions between the histone core and the DNA, and the conformations, positions, and DNA interactions of the histone tails. Understanding chromatin dynamics requires studying all these features at the highest possible resolution. For this, molecular dynamics simulations can be used as a powerful complement or alternative to experimental approaches, from which it is often very challenging to characterize the structural features and atomic interactions controlling nucleosome motions. Molecular dynamics simulations can be performed at different resolutions, by coarse graining the molecular system with varying levels of details. Here we review the successes and the remaining challenges of the application of atomic resolution simulations to study the structure and dynamics of nucleosomes and their complexes with interacting partners.


Subject(s)
DNA/chemistry , Histones/chemistry , Nucleosomes/ultrastructure , Protein Processing, Post-Translational , Acetylation , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , Histones/genetics , Histones/metabolism , Methylation , Molecular Dynamics Simulation , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
8.
Annu Rev Phys Chem ; 71: 101-119, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32017651

ABSTRACT

Chromatosomes are fundamental units of chromatin structure that are formed when a linker histone protein binds to a nucleosome. The positioning of the linker histone on the nucleosome influences the packing of chromatin. Recent simulations and experiments have shown that chromatosomes adopt an ensemble of structures that differ in the geometry of the linker histone-nucleosome interaction. In this article we review the application of Brownian, Monte Carlo, and molecular dynamics simulations to predict the structure of linker histone-nucleosome complexes, to study the binding mechanisms involved, and to predict how this binding affects chromatin fiber structure. These simulations have revealed the sensitivityof the chromatosome structure to variations in DNA and linker histone sequence, as well as to posttranslational modifications, thereby explaining the structural variability observed in experiments. We propose that a concerted application of experimental and computational approaches will reveal the determinants of chromatosome structural variability and how it impacts chromatin packing.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Nucleosomes/metabolism , Animals , Chickens , Chromatin/chemistry , DNA/chemistry , DNA/metabolism , Histones/chemistry , Molecular Dynamics Simulation , Monte Carlo Method , Nucleosomes/chemistry
9.
Biophys J ; 118(9): 2280-2296, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32027821

ABSTRACT

Transcription factor (TF) proteins bind to DNA to regulate gene expression. Normally, accessibility to DNA is required for their function. However, in the nucleus, the DNA is often inaccessible, wrapped around histone proteins in nucleosomes forming the chromatin. Pioneer TFs are thought to induce chromatin opening by recognizing their DNA binding sites on nucleosomes. For example, Oct4, a master regulator and inducer of stem cell pluripotency, binds to DNA in nucleosomes in a sequence-specific manner. Here, we reveal the structural dynamics of nucleosomes that mediate Oct4 binding from molecular dynamics simulations. Nucleosome flexibility and the amplitude of nucleosome motions such as breathing and twisting are enhanced in nucleosomes with multiple TF binding sites. Moreover, the regions around the binding sites display higher local structural flexibility. Probing different structures of Oct4-nucleosome complexes, we show that alternative configurations in which Oct4 recognizes partial binding sites display stable TF-DNA interactions similar to those observed in complexes with free DNA and compatible with the DNA curvature and DNA-histone interactions. Therefore, we propose a structural basis for nucleosome recognition by a pioneer TF that is essential for understanding how chromatin is unraveled during cell fate conversions.


Subject(s)
DNA , Nucleosomes , Binding Sites , Chromatin , Histones/metabolism
10.
FEBS J ; 287(1): 122-144, 2020 01.
Article in English | MEDLINE | ID: mdl-31569299

ABSTRACT

The functional consequences of cancer-associated missense mutations are unclear for the majority of proteins. We have previously demonstrated that the activity of SOX and Pit-Oct-Unc (POU) family factors during pluripotency reprogramming can be switched and enhanced with rationally placed point mutations. Here, we interrogated cancer mutation databases and identified recurrently mutated positions at critical structural interfaces of the DNA-binding domains of paralogous SOX and POU family transcription factors. Using the conversion of mouse embryonic fibroblasts to induced pluripotent stem cells as functional readout, we identified several gain-of-function mutations that enhance pluripotency reprogramming by SOX2 and OCT4. Wild-type SOX17 cannot support reprogramming but the recurrent missense mutation SOX17-V118M is capable of inducing pluripotency. Furthermore, SOX17-V118M promotes oncogenic transformation, enhances thermostability and elevates cellular protein levels of SOX17. We conclude that the mutational profile of SOX and POU family factors in cancer can guide the design of high-performance reprogramming factors. Furthermore, we propose cellular reprogramming as a suitable assay to study the functional impact of cancer-associated mutations.


Subject(s)
Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Mutation, Missense , Neoplasms/pathology , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , SOXF Transcription Factors/genetics , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neoplasms/genetics , Neoplasms/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , SOXF Transcription Factors/metabolism
11.
J Cell Sci ; 132(9)2019 05 08.
Article in English | MEDLINE | ID: mdl-30926623

ABSTRACT

Clathrin-mediated endocytosis (CME) engages over 30 proteins to secure efficient cargo and membrane uptake. While the function of most core CME components is well established, auxiliary mechanisms crucial for fine-tuning and adaptation remain largely elusive. In this study, we identify ArhGEF37, a currently uncharacterized protein, as a constituent of CME. Structure prediction together with quantitative cellular and biochemical studies present a unique BAR domain and PI(4,5)P2-dependent protein-membrane interactions. Functional characterization yields accumulation of ArhGEF37 at dynamin 2-rich late endocytic sites and increased endocytosis rates in the presence of ArhGEF37. Together, these results introduce ArhGEF37 as a regulatory protein involved in endocytosis.


Subject(s)
Dynamin II/metabolism , Endocytosis/physiology , Rho Guanine Nucleotide Exchange Factors , Animals , Clathrin-Coated Vesicles/metabolism , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/metabolism
12.
Structure ; 26(8): 1050-1057, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29937356

ABSTRACT

There is renewed interest in linker histone (LH)-nucleosome binding and how LHs influence eukaryotic DNA compaction. For a long time, the goal was to uncover "the structure of the chromatosome," but recent studies of LH-nucleosome complexes have revealed an ensemble of structures. Notably, the reconstituted LH-nucleosome complexes used in experiments rarely correspond to the sequence combinations present in organisms. For a full understanding of the determinants of the distribution of the chromatosome structural ensemble, studies must include a complete description of the sequences and experimental conditions used, and be designed to enable systematic evaluation of sequence and environmental effects.


Subject(s)
DNA/chemistry , Histones/chemistry , Models, Molecular , Nucleosomes/ultrastructure , Animals , Chickens/genetics , Chickens/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Histones/genetics , Histones/metabolism , Humans , Nucleic Acid Conformation , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Xenopus laevis/genetics , Xenopus laevis/metabolism
13.
Biophys J ; 114(10): 2363-2375, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29759374

ABSTRACT

Linker histone (LH) proteins play a key role in higher-order structuring of chromatin for the packing of DNA in eukaryotic cells and in the regulation of genomic function. The common fruit fly (Drosophila melanogaster) has a single somatic isoform of the LH (H1). It is thus a useful model organism for investigating the effects of the LH on nucleosome compaction and the structure of the chromatosome, the complex formed by binding of an LH to a nucleosome. The structural and mechanistic details of how LH proteins bind to nucleosomes are debated. Here, we apply Brownian dynamics simulations to compare the nucleosome binding of the globular domain of D. melanogaster H1 (gH1) and the corresponding chicken (Gallus gallus) LH isoform, gH5, to identify residues in the LH that critically affect the structure of the chromatosome. Moreover, we investigate the effects of posttranslational modifications on the gH1 binding mode. We find that certain single-point mutations and posttranslational modifications of the LH proteins can significantly affect chromatosome structure. These findings indicate that even subtle differences in LH sequence can significantly shift the chromatosome structural ensemble and thus have implications for chromatin structure and transcriptional regulation.


Subject(s)
Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Chickens , Drosophila melanogaster , Molecular Dynamics Simulation , Nucleic Acid Conformation , Protein Domains
14.
Nucleic Acids Res ; 46(11): 5470-5486, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29669022

ABSTRACT

FOXA1 is a transcription factor capable to bind silenced chromatin to direct context-dependent cell fate conversion. Here, we demonstrate that a compact palindromic DNA element (termed 'DIV' for its diverging half-sites) induces the homodimerization of FOXA1 with strongly positive cooperativity. Alternative structural models are consistent with either an indirect DNA-mediated cooperativity or a direct protein-protein interaction. The cooperative homodimer formation is strictly constrained by precise half-site spacing. Re-analysis of chromatin immunoprecipitation sequencing data indicates that the DIV is effectively targeted by FOXA1 in the context of chromatin. Reporter assays show that FOXA1-dependent transcriptional activity declines when homodimeric binding is disrupted. In response to phosphatidylinositol-3 kinase inhibition DIV sites pre-bound by FOXA1 such as at the PVT1/MYC locus exhibit a strong increase in accessibility suggesting a role of the DIV configuration in the chromatin closed-open dynamics. Moreover, several disease-associated single nucleotide polymorphisms map to DIV elements and show allelic differences in FOXA1 homodimerization, reporter gene expression and are annotated as quantitative trait loci. This includes the rs541455835 variant at the MAPT locus encoding the Tau protein associated with Parkinson's disease. Collectively, the DIV guides chromatin engagement and regulation by FOXA1 and its perturbation could be linked to disease etiologies.


Subject(s)
DNA/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Inverted Repeat Sequences/genetics , Cell Line, Tumor , Chromatin/metabolism , Dimerization , HCT116 Cells , Humans , MCF-7 Cells , Phosphoinositide-3 Kinase Inhibitors , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Thiazoles/pharmacology
15.
J Mol Biol ; 429(23): 3626-3634, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29050852

ABSTRACT

Sox2 and Pax6 co-regulate genes in neural lineages and the lens by forming a ternary complex likely facilitated allosterically through DNA. We used the quantitative and scalable cooperativity-by-sequencing (Coop-seq) approach to interrogate Sox2/Pax6 dimerization on a DNA library where five positions of the Pax6 half-site were randomized yielding 1024 cooperativity factors. Consensus positions normally required for the high-affinity DNA binding by Pax6 need to be mutated for effective dimerization with Sox2. Out of the five randomized bases, a 5' thymidine is present in most of the top ranking elements. However, this thymidine maps to a region outside of the Pax half site and is not expected to directly interact with Pax6 in known binding modes suggesting structural reconfigurations. Re-analysis of ChIP-seq data identified several genomic regions where the cooperativity promoting sequence pattern is co-bound by Sox2 and Pax6. A highly conserved Sox2/Pax6 bound site near the Sprouty2 locus was verified to promote cooperative dimerization designating Sprouty2 as a potential target reliant on Sox2/Pax6 cooperativity in several neural cell types. Collectively, the functional interplay of Sox2 and Pax6 demands the relaxation of high-affinity binding sites and is enabled by alternative DNA sequences. We conclude that this binding mode evolved to warrant that a subset of target genes is only regulated in the presence of suitable partner factors.


Subject(s)
DNA/metabolism , PAX6 Transcription Factor/metabolism , SOXB1 Transcription Factors/metabolism , Sequence Analysis, DNA/methods , DNA/chemistry , DNA/genetics , Humans , Models, Molecular , PAX6 Transcription Factor/chemistry , PAX6 Transcription Factor/genetics , Protein Binding , Protein Conformation , Protein Multimerization , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/genetics
16.
EMBO Rep ; 18(2): 319-333, 2017 02.
Article in English | MEDLINE | ID: mdl-28007765

ABSTRACT

The transcription factor Oct4 is a core component of molecular cocktails inducing pluripotent stem cells (iPSCs), while other members of the POU family cannot replace Oct4 with comparable efficiency. Rather, group III POU factors such as Oct6 induce neural lineages. Here, we sought to identify molecular features determining the differential DNA-binding and reprogramming activity of Oct4 and Oct6. In enhancers of pluripotency genes, Oct4 cooperates with Sox2 on heterodimeric SoxOct elements. By re-analyzing ChIP-Seq data and performing dimerization assays, we found that Oct6 homodimerizes on palindromic OctOct more cooperatively and more stably than Oct4. Using structural and biochemical analyses, we identified a single amino acid directing binding to the respective DNA elements. A change in this amino acid decreases the ability of Oct4 to generate iPSCs, while the reverse mutation in Oct6 does not augment its reprogramming activity. Yet, with two additional amino acid exchanges, Oct6 acquires the ability to generate iPSCs and maintain pluripotency. Together, we demonstrate that cell type-specific POU factor function is determined by select residues that affect DNA-dependent dimerization.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , POU Domain Factors/chemistry , POU Domain Factors/metabolism , Protein Multimerization , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Embryonic Stem Cells , Enhancer Elements, Genetic , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Molecular , Nucleotide Motifs , Octamer Transcription Factors/chemistry , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , POU Domain Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Protein Stability , Transcriptome
17.
Nucleic Acids Res ; 44(14): 6599-613, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27270081

ABSTRACT

Linker histones are essential for DNA compaction in chromatin. They bind to nucleosomes in a 1:1 ratio forming chromatosomes. Alternative configurations have been proposed in which the globular domain of the linker histone H5 (gH5) is positioned either on- or off-dyad between the nucleosomal and linker DNAs. However, the dynamic pathways of chromatosome assembly remain elusive. Here, we studied the conformational plasticity of gH5 in unbound and off-dyad nucleosome-bound forms with classical and accelerated molecular dynamics simulations. We find that the unbound gH5 converts between open and closed conformations, preferring the closed form. However, the open gH5 contributes to a more rigid chromatosome and restricts the motion of the nearby linker DNA through hydrophobic interactions with thymidines. Moreover, the closed gH5 opens and reorients in accelerated simulations of the chromatosome. Brownian dynamics simulations of chromatosome assembly, accounting for a range of amplitudes of nucleosome opening and different nucleosome DNA sequences, support the existence of both on- and off-dyad binding modes of gH5 and reveal alternative, sequence and conformation-dependent chromatosome configurations. Taken together, these findings suggest that the conformational dynamics of linker histones and nucleosomes facilitate alternative chromatosome configurations through an interplay between induced fit and conformational selection.


Subject(s)
Histones/chemistry , Histones/metabolism , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , DNA/chemistry , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Thymidine/metabolism
18.
Biochim Biophys Acta ; 1860(1 Pt A): 67-78, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26493722

ABSTRACT

BACKGROUND: Cytochrome P450 sterol 14α-demethylase (CYP51) is an essential enzyme for sterol biosynthesis and a target for anti-parasitic drug design. However, the design of parasite-specific drugs that inhibit parasitic CYP51 without severe side effects remains challenging. The active site of CYP51 is situated in the interior of the protein. Here, we characterize the potential ligand egress routes and mechanisms in Trypanosoma brucei and human CYP51 enzymes. METHODS: We performed Random Acceleration Molecular Dynamics simulations of the egress of four different ligands from the active site of models of soluble and membrane-bound T. brucei CYP51 and of soluble human CYP51. RESULTS: In the simulations, tunnel 2f, which leads to the membrane, was found to be the predominant ligand egress tunnel for all the ligands studied. Tunnels S, 1 and W, which lead to the cytosol, were also used in T. brucei CYP51, whereas tunnel 1 was the only other tunnel used significantly in human CYP51. The common tunnels found previously in other CYPs were barely used. The ligand egress times were shorter for human than T. brucei CYP51, suggesting lower barriers to ligand passage. Two gating residues, F105 and M460, in T. brucei CYP51 that modulate the opening of tunnels 2f and S were identified. CONCLUSIONS: Although the main egress tunnel was the same, differences in the tunnel-lining residues, ligand passage and tunnel usage were found between T. brucei and human CYP51s. GENERAL SIGNIFICANCE: The results provide a basis for the design of selective anti-parasitic agents targeting the ligand tunnels.


Subject(s)
Drug Design , Sterol 14-Demethylase/chemistry , Trypanosoma brucei brucei/drug effects , Binding Sites , Humans , Ligands , Molecular Dynamics Simulation
19.
Sci Rep ; 5: 13533, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26314899

ABSTRACT

The transcription factors OCT4 and SOX2 are required for generating induced pluripotent stem cells (iPSCs) and for maintaining embryonic stem cells (ESCs). OCT4 and SOX2 associate and bind to DNA in different configurations depending on the arrangement of their individual DNA binding elements. Here we have investigated the role of the different OCT4-SOX2-DNA assemblies in regulating and inducing pluripotency. To this end, we have generated SOX2 mutants that interfere with specific OCT4-SOX2 heterodimer configurations and assessed their ability to generate iPSCs and to rescue ESC self-renewal. Our results demonstrate that the OCT4-SOX2 configuration that dimerizes on a Hoxb1-like composite, a canonical element with juxtaposed individual binding sites, plays a more critical role in the induction and maintenance of pluripotency than any other OCT4-SOX2 configuration. Overall, the results of this study provide new insight into the protein interactions required to establish a de novo pluripotent network and to maintain a true pluripotent cell fate.


Subject(s)
Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Protein Multimerization , SOXB1 Transcription Factors/metabolism , Animals , Cell Differentiation , Cellular Reprogramming , Human Embryonic Stem Cells , Humans , Mice, Transgenic , Models, Molecular , Pluripotent Stem Cells/cytology
20.
PLoS Comput Biol ; 11(6): e1004287, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26067358

ABSTRACT

Highly specific transcriptional regulation depends on the cooperative association of transcription factors into enhanceosomes. Usually, their DNA-binding cooperativity originates from either direct interactions or DNA-mediated allostery. Here, we performed unbiased molecular simulations followed by simulations of protein-DNA unbinding and free energy profiling to study the cooperative DNA recognition by OCT4 and SOX2, key components of enhanceosomes in pluripotent cells. We found that SOX2 influences the orientation and dynamics of the DNA-bound configuration of OCT4. In addition SOX2 modifies the unbinding free energy profiles of both DNA-binding domains of OCT4, the POU specific and POU homeodomain, despite interacting directly only with the first. Thus, we demonstrate that the OCT4-SOX2 cooperativity is modulated by an interplay between protein-protein interactions and DNA-mediated allostery. Further, we estimated the change in OCT4-DNA binding free energy due to the cooperativity with SOX2, observed a good agreement with experimental measurements, and found that SOX2 affects the relative DNA-binding strength of the two OCT4 domains. Based on these findings, we propose that available interaction partners in different biological contexts modulate the DNA exploration routes of multi-domain transcription factors such as OCT4. We consider the OCT4-SOX2 cooperativity as a paradigm of how specificity of transcriptional regulation is achieved through concerted modulation of protein-DNA recognition by different types of interactions.


Subject(s)
DNA/chemistry , DNA/metabolism , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/metabolism , Allosteric Regulation , Molecular Dynamics Simulation , Pluripotent Stem Cells , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL