Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Biochemistry ; 2022 May 17.
Article in English | MEDLINE | ID: mdl-35579944

ABSTRACT

Coactivator-associated arginine methyltransferase 1 (CARM1) is a member of the family of protein arginine methyltransferases. CARM1 catalyzes methyl group transfer from the cofactor S-adenosyl-l-methionine (AdoMet) to both histone and nonhistone protein substrates. CARM1 is involved in a range of cellular processes, mainly involving RNA transcription and gene regulation. As the aberrant expression of CARM1 has been linked to tumorigenesis, the enzyme is a potential therapeutic target, leading to the development of inhibitors and tool compounds engaging with CARM1. To evaluate the effects of these compounds on the activity of CARM1, sensitive and specific analytical methods are needed. While different methods are currently available to assess the activity of methyltransferases, these assays mainly focus on either the measurement of the cofactor product S-adenosyl-l-homocysteine (AdoHcy) or employ radioactive or expensive reagents, each with their own advantages and limitations. To complement the tools currently available for the analysis of CARM1 activity, we here describe the development of a convenient assay employing peptide substrates derived from poly(A)-binding protein 1 (PABP1). This operationally straightforward liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based approach allows for the direct detection of substrate methylation with minimal workup. The method was validated, and its value in characterizing CARM1 activity and inhibition was demonstrated through a comparative analysis involving a set of established small molecules and peptide-based CARM1 inhibitors.

2.
J Med Chem ; 65(17): 11574-11606, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35482954

ABSTRACT

Protein arginine methyltransferases (PRMTs) are important therapeutic targets, playing a crucial role in the regulation of many cellular processes and being linked to many diseases. Yet, there is still much to be understood regarding their functions and the biological pathways in which they are involved, as well as on the structural requirements that could drive the development of selective modulators of PRMT activity. Here we report a deconstruction-reconstruction approach that, starting from a series of type I PRMT inhibitors previously identified by us, allowed for the identification of potent and selective inhibitors of PRMT4, which regardless of the low cell permeability show an evident reduction of arginine methylation levels in MCF7 cells and a marked reduction of proliferation. We also report crystal structures with various PRMTs supporting the observed specificity and selectivity.


Subject(s)
Arginine , Protein-Arginine N-Methyltransferases , Arginine/metabolism , Enzyme Inhibitors/chemistry , Methylation , Protein Processing, Post-Translational
3.
Methods Mol Biol ; 2406: 281-317, 2022.
Article in English | MEDLINE | ID: mdl-35089564

ABSTRACT

Rapid preparation of proteins for functional and structural analysis is a major challenge both in academia and industry. The number potential targets continuously increases and many are difficult to express proteins which, when produced in bacteria, result in insoluble and/or misfolded recombinant proteins, protein aggregates, or unusable low protein yield. We focus here on the baculovirus expression vector system which is now commonly used for heterologous production of human targets. This chapter describes simple and cost-effective protocols that enable iterative cycles of construct design, expression screening and optimization of protein production. We detail time- and cost-effective methods for generation of baculoviruses by homologous recombination and titer evaluation. Handling of insect cell cultures and preparation of bacmid for cotransfection are also presented.


Subject(s)
Baculoviridae , Genetic Vectors , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cell Culture Techniques , Genetic Vectors/genetics , Humans , Insecta/genetics , Insecta/metabolism , Recombinant Proteins/metabolism
4.
Life (Basel) ; 11(11)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34833139

ABSTRACT

PRMT2 belongs to the protein arginine methyltransferase (PRMT) family, which catalyzes the arginine methylation of target proteins. As a type I enzyme, PRMT2 produces asymmetric dimethyl arginine and has been shown to have weak methyltransferase activity on histone substrates in vitro, suggesting that its authentic substrates have not yet been found. PRMT2 contains the canonical PRMT methylation core and a unique Src homology 3 domain. Studies have demonstrated its clear implication in many different cellular processes. PRMT2 acts as a coactivator of several nuclear hormone receptors and is known to interact with a multitude of splicing-related proteins. Furthermore, PRMT2 is aberrantly expressed in several cancer types, including breast cancer and glioblastoma. These reports highlight the crucial role played by PRMT2 and the need for a better characterization of its activity and cellular functions.

5.
Chembiochem ; 22(24): 3469-3476, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34569136

ABSTRACT

The dynamic interplay of post-translational modifications (PTMs) in chromatin provides a communication system for the regulation of gene expression. An increasing number of studies have highlighted the role that such crosstalk between PTMs plays in chromatin recognition. In this study, (bio)chemical and structural approaches were applied to specifically probe the impact of acetylation of Lys18 in the histone H3 tail peptide on peptide recognition by the protein methyltransferase coactivator-associated arginine methyltransferase 1 (CARM1). Peptidomimetics that recapitulate the transition state of protein arginine N-methyltransferases, were designed based on the H3 peptide wherein the target Arg17 was flanked by either a free or an acetylated lysine. Structural studies with these peptidomimetics and the catalytic domain of CARM1 provide new insights into the binding of the H3 peptide within the enzyme active site. While the co-crystal structures reveal that lysine acetylation results in minor conformational differences for both CARM1 and the H3 peptide, acetylation of Lys18 does lead to additional interactions (Van der Waals and hydrogen bonding) and likely reduces the cost of desolvation upon binding, resulting in increased affinity. Informed by these findings a series of smaller peptidomimetics were also prepared and found to maintain potent and selective CARM1 inhibition. These findings provide new insights both into the mechanism of crosstalk between arginine methylation and lysine acetylation as well as towards the development of peptidomimetic CARM1 inhibitors.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Lysine/antagonists & inhibitors , Peptidomimetics/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Acetylation , Animals , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Lysine/metabolism , Mice , Models, Molecular , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Protein Conformation , Protein-Arginine N-Methyltransferases/metabolism , Substrate Specificity
6.
Article in English | MEDLINE | ID: mdl-29685976

ABSTRACT

DNA, RNA and histone methylation is implicated in various human diseases such as cancer or viral infections, playing a major role in cell process regulation, especially in modulation of gene expression. Here we developed a convergent synthetic pathway starting from a protected bromomethylcytosine derivative to synthesize transition state analogues of the DNA methyltransferases. This approach led to seven 5-methylcytosine-adenosine compounds that were, surprisingly, inactive against hDNMT1, hDNMT3Acat, TRDMT1 and other RNA human and viral methyltransferases. Interestingly, compound 4 and its derivative 2 showed an inhibitory activity against PRMT4 in the micromolar range. Crystal structures showed that compound 4 binds to the PRMT4 active site, displacing strongly the S-adenosyl-l-methionine cofactor, occupying its binding site, and interacting with the arginine substrate site through the cytosine moiety that probes the space filled by a substrate peptide methylation intermediate. Furthermore, the binding of the compounds induces important structural switches. These findings open new routes for the conception of new potent PRMT4 inhibitors based on the 5-methylcytosine-adenosine scaffold.This article is part of a discussion meeting issue 'Frontiers in epigenetic chemical biology'.


Subject(s)
Catalytic Domain , Methyltransferases/chemical synthesis , Peptides/metabolism , Humans
7.
Nucleic Acids Res ; 45(9): 5399-5413, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28115638

ABSTRACT

Selenoprotein synthesis requires the co-translational recoding of a UGASec codon. This process involves an RNA structural element, called Selenocysteine Insertion Sequence (SECIS) and the SECIS binding protein 2 (SBP2). Several selenoprotein mRNAs undergo unusual cap hypermethylation by the trimethylguanosine synthase 1 (Tgs1), which is recruited by the ubiquitous Survival of MotoNeurons (SMN) protein. SMN, the protein involved in spinal muscular atrophy, is part of a chaperone complex that collaborates with the methylosome for RNP assembly. Here, we analyze the role of individual SMN and methylosome components in selenoprotein mRNP assembly and translation. We show that SBP2 interacts directly with four proteins of the SMN complex and the methylosome core proteins. Nevertheless, SBP2 is not a methylation substrate of the methylosome. We found that both SMN and methylosome complexes are required for efficient translation of the selenoprotein GPx1 in vivo. We establish that the steady-state level of several selenoprotein mRNAs, major regulators of oxidative stress damage in neurons, is specifically reduced in the spinal cord of SMN-deficient mice and that cap hypermethylation of GPx1 mRNA is affected. Altogether we identified a new function of the SMN complex and the methylosome in selenoprotein mRNP assembly and expression.


Subject(s)
Protein Biosynthesis , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , SMN Complex Proteins/metabolism , Selenoproteins/metabolism , Glutathione Peroxidase , HEK293 Cells , HeLa Cells , Humans , Methylation , Models, Biological , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Protein Binding , Spinal Cord/metabolism , Glutathione Peroxidase GPX1
8.
FEBS J ; 284(1): 77-96, 2017 01.
Article in English | MEDLINE | ID: mdl-27879050

ABSTRACT

PRMT2 is the less-characterized member of the protein arginine methyltransferase family in terms of structure, activity, and cellular functions. PRMT2 is a modular protein containing a catalytic Ado-Met-binding domain and unique Src homology 3 domain that binds proteins with proline-rich motifs. PRMT2 is involved in a variety of cellular processes and has diverse roles in transcriptional regulation through different mechanisms depending on its binding partners. PRMT2 has been demonstrated to have weak methyltransferase activity on a histone H4 substrate, but its optimal substrates have not yet been identified. To obtain insights into the function and activity of PRMT2, we solve several crystal structures of PRMT2 from two homologs (zebrafish and mouse) in complex with either the methylation product S-adenosyl-L-homocysteine or other compounds including the first synthetic PRMT2 inhibitor (Cp1) studied so far. We reveal that the N-terminal-containing SH3 module is disordered in the full-length crystal structures, and highlights idiosyncratic features of the PRMT2 active site. We identify a new nonhistone protein substrate belonging to the serine-/arginine-rich protein family which interacts with PRMT2 and we characterize six methylation sites by mass spectrometry. To better understand structural basis for Cp1 binding, we also solve the structure of the complex PRMT4:Cp1. We compare the inhibitor-protein interactions occurring in the PRMT2 and PRMT4 complex crystal structures and show that this compound inhibits efficiently PRMT2. These results are a first step toward a better understanding of PRMT2 substrate recognition and may accelerate the development of structure-based drug design of PRMT2 inhibitors. DATABASE: All coordinates and structure factors have been deposited in the Protein Data Bank: zPRMT21-408 -SFG = 5g02; zPRMT273-408 -SAH = 5fub; mPRMT21-445 -SAH = 5ful; mPRMT21-445 -Cp1 = 5fwa, mCARM1130-487 -Cp1 = 5k8v.


Subject(s)
Enzyme Inhibitors/chemistry , Protein-Arginine N-Methyltransferases/chemistry , S-Adenosylhomocysteine/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Methylation , Mice , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Zebrafish
9.
Sci Rep ; 6: 19725, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813996

ABSTRACT

Translationally Controlled Tumor Protein (TCTP) is anti-apoptotic, key in development and cancer, however without the typical Bcl2 family members' structure. Here we report that TCTP contains a BH3-like domain and forms heterocomplexes with Bcl-xL. The crystal structure of a Bcl-xL deletion variant-TCTP11-31 complex reveals that TCTP refolds in a helical conformation upon binding the BH3-groove of Bcl-xL, although lacking the h1-subregion interaction. Experiments using in vitro-vivo reconstituted systems and TCTP(+/-) mice indicate that TCTP activates the anti-apoptotic function of Bcl-xL, in contrast to all other BH3-proteins. Replacing the non-conserved h1 of TCTP by that of Bax drastically increases the affinity of this hybrid for Bcl-xL, modifying its biological properties. This work reveals a novel class of BH3-proteins potentiating the anti-apoptotic function of Bcl-xL.


Subject(s)
Biomarkers, Tumor/metabolism , Protein Interaction Domains and Motifs , bcl-X Protein/metabolism , Amino Acid Sequence , Animals , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Biomarkers, Tumor/chemistry , Cell Membrane Permeability , Mice , Models, Molecular , Multiprotein Complexes/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Tumor Protein, Translationally-Controlled 1 , bcl-2-Associated X Protein/metabolism , bcl-X Protein/chemistry
10.
J Struct Biol ; 191(2): 175-83, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26094878

ABSTRACT

PRMT6 is a protein arginine methyltransferase involved in transcriptional regulation, human immunodeficiency virus pathogenesis, DNA base excision repair, and cell cycle progression. Like other PRMTs, PRMT6 is overexpressed in several cancer types and is therefore considered as a potential anti-cancer drug target. In the present study, we described six crystal structures of PRMT6 from Mus musculus, solved and refined at 1.34 Å for the highest resolution structure. The crystal structures revealed that the folding of the helix αX is required to stabilize a productive active site before methylation of the bound peptide can occur. In the absence of cofactor, metal cations can be found in the catalytic pocket at the expected position of the guanidinium moiety of the target arginine substrate. Using mass spectrometry under native conditions, we show that PRMT6 dimer binds two cofactor and a single H4 peptide molecules. Finally, we characterized a new site of in vitro automethylation of mouse PRMT6 at position 7.


Subject(s)
Protein-Arginine N-Methyltransferases/chemistry , Amino Acid Sequence , Animals , Cloning, Molecular , Crystallography, X-Ray , Mass Spectrometry , Methylation , Mice , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/physiology , Sequence Alignment , Structure-Activity Relationship
11.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 9): 2401-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25195753

ABSTRACT

Protein arginine methyltransferase 7 (PRMT7) is a type III arginine methyltransferase which has been implicated in several biological processes such as transcriptional regulation, DNA damage repair, RNA splicing, cell differentiation and metastasis. PRMT7 is a unique but less characterized member of the family of PRMTs. The crystal structure of full-length PRMT7 from Mus musculus refined at 1.7 Šresolution is described. The PRMT7 structure is composed of two catalytic modules in tandem forming a pseudo-dimer and contains only one AdoHcy molecule bound to the N-terminal module. The high-resolution crystal structure presented here revealed several structural features showing that the second active site is frozen in an inactive state by a conserved zinc finger located at the junction between the two PRMT modules and by the collapse of two degenerated AdoMet-binding loops.


Subject(s)
Arginine/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Zinc Fingers , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Cloning, Molecular , Dimerization , Methylation , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Sequence Homology, Amino Acid
12.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 1): 80-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24419624

ABSTRACT

Protein arginine methyltransferase 7 (PRMT7) is a unique but less characterized member of the family of protein arginine methyltransferases (PRMTs) that plays a role in male germline gene imprinting. PRMT7 is the only known PRMT member that catalyzes the monomethylation but not the dimethylation of the target arginine residues and harbours two catalytic domains in tandem. PRMT7 genes from five different species were cloned and expressed in Escherichia coli and Sf21 insect cells. Four gave soluble proteins from Sf21 cells, of which two were homogeneous and one gave crystals. The mouse PRMT7 structure was solved by the single anomalous dispersion method using a crystal soaked with thimerosal that diffracted to beyond 2.1 Šresolution. The crystal belonged to space group P4(3)2(1)2, with unit-cell parameters a = b = 97.4, c = 168.1 Šand one PRMT7 monomer in the asymmetric unit. The structure of another crystal form belonging to space group I222 was solved by molecular replacement.


Subject(s)
Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/isolation & purification , Amino Acid Sequence , Animals , Arabidopsis Proteins/metabolism , Chromatography, Gel , Cloning, Molecular , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Humans , Light , Male , Mice , Molecular Sequence Data , Scattering, Radiation , Transfection
13.
PLoS Pathog ; 9(9): e1003645, 2013.
Article in English | MEDLINE | ID: mdl-24086136

ABSTRACT

The treatment of schistosomiasis, a disease caused by blood flukes parasites of the Schistosoma genus, depends on the intensive use of a single drug, praziquantel, which increases the likelihood of the development of drug-resistant parasite strains and renders the search for new drugs a strategic priority. Currently, inhibitors of human epigenetic enzymes are actively investigated as novel anti-cancer drugs and have the potential to be used as new anti-parasitic agents. Here, we report that Schistosoma mansoni histone deacetylase 8 (smHDAC8), the most expressed class I HDAC isotype in this organism, is a functional acetyl-L-lysine deacetylase that plays an important role in parasite infectivity. The crystal structure of smHDAC8 shows that this enzyme adopts a canonical α/ß HDAC fold, with specific solvent exposed loops corresponding to insertions in the schistosome HDAC8 sequence. Importantly, structures of smHDAC8 in complex with generic HDAC inhibitors revealed specific structural changes in the smHDAC8 active site that cannot be accommodated by human HDACs. Using a structure-based approach, we identified several small-molecule inhibitors that build on these specificities. These molecules exhibit an inhibitory effect on smHDAC8 but show reduced affinity for human HDACs. Crucially, we show that a newly identified smHDAC8 inhibitor has the capacity to induce apoptosis and mortality in schistosomes. Taken together, our biological and structural findings define the framework for the rational design of small-molecule inhibitors specifically interfering with schistosome epigenetic mechanisms, and further support an anti-parasitic epigenome targeting strategy to treat neglected diseases caused by eukaryotic pathogens.


Subject(s)
Epigenesis, Genetic , Helminth Proteins/chemistry , Histone Deacetylases/chemistry , Protein Folding , Schistosoma mansoni/enzymology , Animals , Helminth Proteins/genetics , Helminth Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Protein Structure, Secondary , Schistosoma mansoni/genetics
14.
Science ; 339(6120): 694-8, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23393263

ABSTRACT

E6 viral oncoproteins are key players in epithelial tumors induced by papillomaviruses in vertebrates, including cervical cancer in humans. E6 proteins target many host proteins by specifically interacting with acidic LxxLL motifs. We solved the crystal structures of bovine (BPV1) and human (HPV16) papillomavirus E6 proteins bound to LxxLL peptides from the focal adhesion protein paxillin and the ubiquitin ligase E6AP, respectively. In both E6 proteins, two zinc domains and a linker helix form a basic-hydrophobic pocket, which captures helical LxxLL motifs in a way compatible with other interaction modes. Mutational inactivation of the LxxLL binding pocket disrupts the oncogenic activities of both E6 proteins. This work reveals the structural basis of both the multifunctionality and the oncogenicity of E6 proteins.


Subject(s)
Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/metabolism , Paxillin/chemistry , Protein Interaction Domains and Motifs , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Amino Acid Motifs , Amino Acid Sequence , Bovine papillomavirus 1 , Crystallography, X-Ray , Human papillomavirus 16 , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Paxillin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Point Mutation , Protein Structure, Secondary , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
15.
J Med Chem ; 55(22): 9875-90, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23095008

ABSTRACT

Arginine methylation is a common post-translational modification that is crucial in modulating gene expression at multiple critical levels. The arginine methyltransferases (PRMTs) are envisaged as promising druggable targets, but their role in physiological and pathological pathways is far from being clear due to the limited number of modulators reported to date. In this effort, enzyme activators can be invaluable tools useful as gain-of-function reagents to interrogate the biological roles in cells and in vivo of PRMTs. Yet the identification of such molecules is rarely pursued. Herein we describe a series of aryl ureido acetamido indole carboxylates (dubbed "uracandolates"), able to increase the methylation of histone (H3) or nonhistone (polyadenylate-binding protein 1, PABP1) substrates induced by coactivator-associated arginine methyltransferase 1 (CARM1), both in in vitro and cellular settings. To the best of our knowledge, this is the first report of compounds acting as CARM1 activators.


Subject(s)
Arginine/genetics , Enzyme Activators/pharmacology , Histones/genetics , Methylation/drug effects , Poly(A)-Binding Protein I/genetics , Protein-Arginine N-Methyltransferases/metabolism , Small Molecule Libraries/pharmacology , Arginine/chemistry , Blotting, Western , Catalysis , Enzyme Activators/chemical synthesis , Humans , Indoles/chemistry , Molecular Structure , Protein Processing, Post-Translational , Structure-Activity Relationship , Surface Plasmon Resonance , Trans-Activators
16.
Proc Natl Acad Sci U S A ; 109(10): E588-94, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22355136

ABSTRACT

Transcription regulation by steroid hormones, vitamin derivatives, and metabolites is mediated by nuclear receptors (NRs), which play an important role in ligand-dependent gene expression and human health. NRs function as homodimers or heterodimers and are involved in a combinatorial, coordinated and sequentially orchestrated exchange between coregulators (corepressors, coactivators). The architecture of DNA-bound functional dimers positions the coregulators proteins. We previously demonstrated that retinoic acid (RAR-RXR) and vitamin D3 receptors (VDR-RXR) heterodimers recruit only one coactivator molecule asymmetrically without steric hindrance for the binding of a second cofactor. We now address the problem of homodimers for which the presence of two identical targets enhances the functional importance of the mode of binding. Using structural and biophysical methods and RAR as a model, we could dissect the molecular mechanism of coactivator recruitment to homodimers. Our study reveals an allosteric mechanism whereby binding of a coactivator promotes formation of nonsymmetrical RAR homodimers with a 21 stoichiometry. Ligand conformation and the cofactor binding site of the unbound receptor are affected through the dimer interface. A similar control mechanism is observed with estrogen receptor (ER) thus validating the negative cooperativity model for an established functional homodimer. Correlation with published data on other NRs confirms the general character of this regulatory pathway.


Subject(s)
Cell Nucleus/metabolism , Allosteric Site , Biophysics/methods , Crystallography, X-Ray/methods , Dimerization , Humans , Kinetics , Ligands , Models, Biological , Models, Molecular , Molecular Conformation , Nuclear Receptor Coactivator 1/chemistry , Peptides/chemistry , Protein Binding , Receptors, Calcitriol/chemistry , Receptors, Retinoic Acid/chemistry
17.
Bioorg Med Chem ; 19(12): 3717-31, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21440447

ABSTRACT

Arginine methylation is an epigenetic modification that receives increasing interest as it plays an important role in several diseases. This is especially true for hormone-dependent cancer, seeing that histone methylation by arginine methyltransferase I (PRMT1) is involved in the activation of sexual hormone receptors. Therefore, PRMT inhibitors are potential drugs and interesting tools for cell biology. A dapsone derivative called allantodapsone previously identified by our group served as a lead structure for inhibitor synthesis. Acylated derivatives of p-aminobenzenesulfonamides and the antilepra drug dapsone were identified as new inhibitors of PRMT1 by in vitro testing. The bis-chloroacetyl amide of dapsone selectively inhibited human PRMT1 in the low micromolar region and was selective for PRMT1 as compared to the arginine methyltransferase CARM1 and the lysine methyltransferase Set7/9. It showed anticancer activity on MCF7a and LNCaP cells and blocked androgen dependent transcription specifically in a reporter gene system. Likewise, a transcriptional block was also demonstrated in LNCaP cells using quantitative RT-PCR on the mRNA of androgen dependent genes.


Subject(s)
Antineoplastic Agents/chemical synthesis , Dapsone/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Methyltransferases/antagonists & inhibitors , Sulfonamides/chemical synthesis , Acylation/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dapsone/chemistry , Dapsone/pharmacology , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins , Models, Molecular , Molecular Structure , Protein-Arginine N-Methyltransferases , Receptors, Androgen/genetics , Sulfonamides/chemistry , Sulfonamides/pharmacology
18.
EMBO J ; 29(23): 3979-91, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21057455

ABSTRACT

Binding of elongation factor Spt6 to Iws1 provides an effective means for coupling eukaryotic mRNA synthesis, chromatin remodelling and mRNA export. We show that an N-terminal region of Spt6 (Spt6N) is responsible for interaction with Iws1. The crystallographic structures of Encephalitozoon cuniculi Iws1 and the Iws1/Spt6N complex reveal two conserved binding subdomains in Iws1. The first subdomain (one HEAT repeat; HEAT subdomain) is a putative phosphoprotein-binding site most likely involved in an Spt6-independent function of Iws1. The second subdomain (two ARM repeats; ARM subdomain) specifically recognizes a bipartite N-terminal region of Spt6. Mutations that alter this region of Spt6 cause severe phenotypes in vivo. Importantly, the ARM subdomain of Iws1 is conserved in several transcription factors, including TFIIS, Elongin A and Med26. We show that the homologous region in yeast TFIIS enables this factor to interact with SAGA and the Mediator subunits Spt8 and Med13, suggesting the molecular basis for TFIIS recruitment at promoters. Taken together, our results provide new structural information about the Iws1/Spt6 complex and reveal a novel interaction domain used for the formation of transcription networks.


Subject(s)
Encephalitozoon cuniculi/chemistry , Fungal Proteins/chemistry , Protein Interaction Domains and Motifs , Transcription Factors/chemistry , Transcriptional Elongation Factors/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Elongin , Encephalitozoon cuniculi/genetics , Encephalitozoon cuniculi/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Phenotype , Point Mutation , Protein Structure, Tertiary , Sequence Alignment , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
19.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 7): 745-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20606254

ABSTRACT

Tfb5 interacts with the Tfb2 subunit of the general transcription factor TFIIH to ensure efficient nucleotide-excision repair in eukaryotes. The crystal structure of the complex between Tfb5 and the C-terminal region of Tfb2 (Tfb2C) from Saccharomyces cerevisiae has recently been reported. Here, the structure-determination process is described as a case study. Although crystals were obtained readily, it was not possible to determine experimental phases from a first crystal form (Tfb2(412-513)-Tfb5(2-72)) that diffracted to 2.6 A resolution. Shortening of the Tfb2C from its N-terminus was decisive and modified the crystal packing, leading to a second crystal form (Tfb2(435-513)-Tfb5(2-72)). These crystals diffracted to 1.7 A resolution with excellent mosaicity and allowed structure determination by conventional approaches using heavy atoms. The refined structure from the second crystal form was used to solve the structure of the first crystal form by molecular replacement. Comparison of the two structures revealed that the N-terminal region of Tfb2C and (to a lesser extent) the C-terminal region of Tfb5 contributed to the crystal packing. A detailed analysis illustrates how variation in domain boundaries influences crystal packing and quality.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Transcription Factor TFIIH/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription Factor TFIIH/metabolism
20.
Article in English | MEDLINE | ID: mdl-18097104

ABSTRACT

The ligand-binding domain (LBD) of human oestrogen receptor alpha was produced in Escherichia coli as a cleavable thioredoxin (Trx) fusion in order to improve solubility. Crystallization trials with either cleaved and purified LBD or with the purified fusion protein both failed to produce crystals. In another attempt, Trx was not removed from the LBD after endoproteolytic cleavage and its presence promoted nucleation and subsequent crystal growth, which allowed the structure determination of two different LBD-ligand-coactivator peptide complexes at 2.3 A resolution. This technique is likely to be applicable to other low-solubility proteins.


Subject(s)
Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/isolation & purification , Thioredoxins/chemistry , Binding Sites , Crystallization , Crystallography, X-Ray , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Humans , Ligands , Polymorphism, Single Nucleotide , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility , Thioredoxins/genetics , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...