Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
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.
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.

4.
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
5.
EMBO J ; 39(23): e104369, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33124732

ABSTRACT

Organelles are physically connected in membrane contact sites. The endoplasmic reticulum possesses three major receptors, VAP-A, VAP-B, and MOSPD2, which interact with proteins at the surface of other organelles to build contacts. VAP-A, VAP-B, and MOSPD2 contain an MSP domain, which binds a motif named FFAT (two phenylalanines in an acidic tract). In this study, we identified a non-conventional FFAT motif where a conserved acidic residue is replaced by a serine/threonine. We show that phosphorylation of this serine/threonine is critical for non-conventional FFAT motifs (named Phospho-FFAT) to be recognized by the MSP domain. Moreover, structural analyses of the MSP domain alone or in complex with conventional and Phospho-FFAT peptides revealed new mechanisms of interaction. Based on these new insights, we produced a novel prediction algorithm, which expands the repertoire of candidate proteins with a Phospho-FFAT that are able to create membrane contact sites. Using a prototypical tethering complex made by STARD3 and VAP, we showed that phosphorylation is instrumental for the formation of ER-endosome contacts, and their sterol transfer function. This study reveals that phosphorylation acts as a general switch for inter-organelle contacts.


Subject(s)
Lipid Metabolism , Membrane Proteins/metabolism , Receptors, Chemokine/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Binding Sites , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Humans , Lipids , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Phosphorylation , Protein Binding , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
6.
Bioorg Chem ; 98: 103750, 2020 05.
Article in English | MEDLINE | ID: mdl-32182520

ABSTRACT

Aminobenzosuberone-based PfA-M1 inhibitors were explored as novel antimalarial agents against two different Plasmodium falciparum strains. The 4-phenyl derivative 7c exhibited the most encouraging growth inhibitory activity with IC50 values of 6.5-11.2 µM. X-ray crystal structures and early assessment of DMPK/ADME-Tox parameters allowed us to initiate structure-based drug design approach and understand the liabilities (such as potential metabolic and aqueous solubility issues) as well as identify the opportunities for improvement of this aminobenzosuberone series. It also suggested that compound 7c should be regarded as an attractive chemical tool to investigate the different biological roles of this multifunctional PfA-M1 protein.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Anisoles/pharmacology , Antimalarials/pharmacology , Cycloheptanes/pharmacology , Enzyme Inhibitors/pharmacology , Plasmodium falciparum/drug effects , Aminopeptidases/metabolism , Anisoles/chemical synthesis , Anisoles/chemistry , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cycloheptanes/chemical synthesis , Cycloheptanes/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Structure-Activity Relationship
7.
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
8.
Proteins ; 85(8): 1413-1421, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28383176

ABSTRACT

Aminopeptidases are ubiquitous hydrolases that cleave the N-terminal residues of proteins and oligopeptides. They are broadly distributed throughout all kingdoms of life and have been implicated in a wide variety of physiological processes, including viral infection, parasite metabolism, protein processing, regulation of peptide hormones, and cancer cell proliferation. Members of the M1 family, also termed gluzincins, are defined by two highly conserved motifs in the catalytic domain: a zinc-binding motif, HEXXH-(X18)-E; and an exopeptidase motif, GXMEN. We report the high-resolution X-ray structures of E. coli aminopeptidase N (PepN) in complex with three aminobenzosuberone scaffolds that display various Ki values (50, 0.33, and 0.034 µM) and provide a compelling view of the outstanding selectivity of these chemical entities for the M1 aminopeptidases. This series of inhibitors interacts as transition state mimics with highly conserved residues of the catalytic machinery and substrate recognition sites. Structural comparisons and model-building studies allowed a deep interpretation of the SAR observed for bacterial, as well as mammalian enzymes. Proteins 2017; 85:1413-1421. © 2017 Wiley Periodicals, Inc.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Anisoles/chemistry , Bacterial Proteins/antagonists & inhibitors , Cycloheptanes/chemistry , Escherichia coli/chemistry , Protease Inhibitors/chemistry , Amino Acid Motifs , Aminopeptidases/chemistry , Aminopeptidases/genetics , Aminopeptidases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Epoxide Hydrolases/chemistry , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Structural Homology, Protein , Substrate Specificity , Thermodynamics
9.
Proc Natl Acad Sci U S A ; 114(14): 3625-3630, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28330993

ABSTRACT

Coactivator associated arginine methyltransferase 1 (CARM1) is a member of the protein arginine methyltransferase (PRMT) family and methylates a range of proteins in eukaryotic cells. Overexpression of CARM1 is implicated in a number of cancers, and it is therefore seen as a potential therapeutic target. Peptide sequences derived from the well-defined CARM1 substrate poly(A)-binding protein 1 (PABP1) were covalently linked to an adenosine moiety as in the AdoMet cofactor to generate transition state mimics. These constructs were found to be potent CARM1 inhibitors and also formed stable complexes with the enzyme. High-resolution crystal structures of CARM1 in complex with these compounds confirm a mode of binding that is indeed reflective of the transition state at the CARM1 active site. Given the transient nature of PRMT-substrate complexes, such transition state mimics represent valuable chemical tools for structural studies aimed at deciphering the regulation of arginine methylation mediated by the family of arginine methyltransferases.


Subject(s)
Peptides/pharmacology , Poly(A)-Binding Protein I/chemistry , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , Adenosine/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Mice , Models, Molecular , Peptides/chemistry , Protein Binding , Protein-Arginine N-Methyltransferases/antagonists & inhibitors
10.
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
11.
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
12.
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
13.
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
14.
FEBS J ; 282(5): 819-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25604895

ABSTRACT

UNLABELLED: Modification of wobble uridines of many eukaryotic tRNAs requires the Elongator complex, a highly conserved six-subunit eukaryotic protein assembly, as well as the Killer toxin-insensitive (Kti) proteins 11-14. Kti11 was additionally shown to be implicated in the biosynthesis of diphthamide, a post-translationally modified histidine of translation elongation factor 2. Recent data indicate that iron-bearing Kti11 functions as an electron donor to the [4Fe-4S] cluster of radical S-Adenosylmethionine enzymes, triggering the subsequent radical reaction. We show here that recombinant yeast Kti11 forms a stable 1 : 1 complex with Kti13. To obtain insights into the function of this heterodimer, the Kti11/Kti13 complex was purified to homogeneity, crystallized, and its structure determined at 1.45 Å resolution. The importance of several residues mediating complex formation was confirmed by mutagenesis. Kti13 adopts a fold characteristic of RCC1-like proteins. The seven-bladed ß-propeller consists of a unique mixture of four- and three-stranded blades. In the complex, Kti13 orients Kti11 and restricts access to its electron-carrying iron atom, constraining the electron transfer capacity of Kti11. Based on these findings, we propose a role for Kti13, and discuss the possible functional implications of complex formation. DATABASE: Structural data have been submitted to the Protein Data Bank under accession number 4X33.


Subject(s)
RNA, Transfer/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Electron Transport , Iron/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , RNA, Transfer/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Static Electricity
15.
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
16.
J Mol Biol ; 426(20): 3442-53, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-24657767

ABSTRACT

Schistosomiasis, caused by the parasitic flatworm Schistosoma mansoni and related species, is a tropical disease that affects over 200 million people worldwide. A new approach for targeting eukaryotic parasites is to tackle their dynamic epigenetic machinery that is necessary for the extensive phenotypic changes during the life cycle of the parasite. Recently, we identified S. mansoni histone deacetylase 8 (smHDAC8) as a potential target for antiparasitic therapy. Here, we present results on the investigations of a focused set of HDAC (histone deacetylase) inhibitors on smHDAC8. Besides several active hydroxamates, we identified a thiol-based inhibitor that inhibited smHDAC8 activity in the micromolar range with unexpected selectivity over the human isotype, which has not been observed so far. The crystal structure of smHDAC8 complexed with the thiol derivative revealed that the inhibitor is accommodated in the catalytic pocket, where it interacts with both the catalytic zinc ion and the essential catalytic tyrosine (Y341) residue via its mercaptoacetamide warhead. To our knowledge, this is the first complex crystal structure of any HDAC inhibited by a mercaptoacetamide inhibitor, and therefore, this finding offers a rationale for further improvement. Finally, an ester prodrug of the thiol HDAC inhibitor exhibited antiparasitic activity on cultured schistosomes in a dose-dependent manner.


Subject(s)
Antiparasitic Agents/chemistry , Helminth Proteins/chemistry , Histone Deacetylases/chemistry , Schistosoma mansoni/enzymology , Thioacetamide/chemistry , Animals , Antiparasitic Agents/metabolism , Antiparasitic Agents/pharmacology , Apoptosis/drug effects , Biocatalysis/drug effects , Crystallography, X-Ray , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Schistosoma mansoni/drug effects , Schistosoma mansoni/physiology , Schistosomiasis mansoni/parasitology , Thioacetamide/metabolism , Thioacetamide/pharmacology , Vorinostat
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL