Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Proteins ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38666764

ABSTRACT

Proteases that recognize linear amino acid sequences with high specificity became indispensable tools of recombinant protein technology for the removal of various fusion tags. Due to its stringent sequence specificity, the catalytic domain of the nuclear inclusion cysteine protease of tobacco etch virus (TEV PR) is also a widely applied reagent for enzymatic removal of fusion tags. For this reason, efforts have been made to improve its stability and modify its specificity. For example, P1' autoproteolytic cleavage-resistant mutant (S219V) TEV PR was found not only to be nearly impervious to self-inactivation, but also exhibited greater stability and catalytic efficiency than the wild-type enzyme. An R203G substitution has been reported to further relax the P1' specificity of the enzyme, however, these results were obtained from crude intracellular assays. Until now, there has been no rigorous comparison of the P1' specificity of the S219V and S219V/R203G mutants in vitro, under carefully controlled conditions. Here, we compare the P1' amino acid preferences of these single and double TEV PR mutants. The in vitro analysis was performed by using recombinant protein substrates representing 20 P1' variants of the consensus TENLYFQ*SGT cleavage site, and synthetic oligopeptide substrates were also applied to study a limited set of the most preferred variants. In addition, the enzyme-substrate interactions were analyzed in silico. The results indicate highly similar P1' preferences for both enzymes, many side-chains can be accommodated by the S1' binding sites, but the kinetic assays revealed lower catalytic efficiency for the S219V/R203G than for the S219V mutant.

2.
J Struct Biol ; 214(4): 107918, 2022 12.
Article in English | MEDLINE | ID: mdl-36343842

ABSTRACT

C/EBPß is a key regulator of numerous cellular processes, but it can also contribute to tumorigenesis and viral diseases. It binds to specific DNA sequences (C/EBP sites) and interacts with other transcription factors to control expression of multiple eukaryotic genes in a tissue and cell-type dependent manner. A body of evidence has established that cell-type-specific regulatory information is contained in the local DNA sequence of the binding motif. In human epithelial cells, C/EBPß is an essential cofactor for TGFß signaling in the case of Smad2/3/4 and FoxO-dependent induction of the cell cycle inhibitor, p15INK4b. In the TGFß-responsive region 2 of the p15INK4b promoter, the Smad binding site is flanked by a C/EBP site, CTTAA•GAAAG, which differs from the canonical, palindromic ATTGC•GCAAT motif. The X-ray crystal structure of C/EBPß bound to the p15INK4b promoter fragment shows how GCGC-to-AAGA substitution generates changes in the intermolecular interactions in the protein-DNA interface that enhances C/EBPß binding specificity, limits possible epigenetic regulation of the promoter, and generates a DNA element with a unique pattern of methyl groups in the major groove. Significantly, CT/GA dinucleotides located at the 5'ends of the double stranded element maintain local narrowing of the DNA minor groove width that is necessary for DNA recognition. Our results suggest that C/EBPß would accept all forms of modified cytosine in the context of the CpT site. This contrasts with the effect on the consensus motif, where C/EBPß binding is modestly increased by cytosine methylation, but substantially decreased by hydroxymethylation.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Epigenesis, Genetic , Humans , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Cycle , Cytosine , DNA/genetics
3.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232729

ABSTRACT

ATP-dependent Lon proteases are key participants in the quality control system that supports the homeostasis of the cellular proteome. Based on their unique structural and biochemical properties, Lon proteases have been assigned in the MEROPS database to three subfamilies (A, B, and C). All Lons are single-chain, multidomain proteins containing an ATPase and protease domains, with different additional elements present in each subfamily. LonA and LonC proteases are soluble cytoplasmic enzymes, whereas LonBs are membrane-bound. Based on an analysis of the available sequences of Lon proteases, we identified a number of enzymes currently assigned to the LonB subfamily that, although presumably membrane-bound, include structural features more similar to their counterparts in the LonA subfamily. This observation was confirmed by the crystal structure of the proteolytic domain of the enzyme previously assigned as Bacillus subtilis LonB, combined with the modeled structure of its ATPase domain. Several structural features present in both domains differ from their counterparts in either LonA or LonB subfamilies. We thus postulate that this enzyme is the founding member of a newly identified LonBA subfamily, so far found only in the gene sequences of firmicutes.


Subject(s)
Protease La , ATP-Dependent Proteases/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Peptide Hydrolases/metabolism , Protease La/genetics , Protease La/metabolism , Proteome/metabolism
4.
Annu Rev Genet ; 47: 405-31, 2013.
Article in English | MEDLINE | ID: mdl-24274754

ABSTRACT

RNase III is a global regulator of gene expression in Escherichia coli that is instrumental in the maturation of ribosomal and other structural RNAs. We examine here how RNase III itself is regulated in response to growth and other environmental changes encountered by the cell and how, by binding or processing double-stranded RNA (dsRNA) intermediates, RNase III controls the expression of genes. Recent insight into the mechanism of dsRNA binding and processing, gained from structural studies of RNase III, is reviewed. Structural studies also reveal new cleavage sites in the enzyme that can generate longer 3' overhangs.


Subject(s)
Ribonuclease III/physiology , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Amino Acid Motifs , Bacteriophage lambda/genetics , Catalysis , Clustered Regularly Interspaced Short Palindromic Repeats , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Eukaryotic Cells/enzymology , Gene Expression Regulation, Bacterial , Nucleic Acid Conformation , Operon , Prokaryotic Cells/enzymology , Protein Processing, Post-Translational , RNA/metabolism , RNA Processing, Post-Transcriptional , RNA, Bacterial/metabolism , RNA, Double-Stranded/metabolism , RNA, Ribosomal/metabolism , RNA, Small Untranslated/genetics , Ribonuclease III/chemistry , Ribonuclease III/classification , Ribonuclease III/genetics , Structure-Activity Relationship , Substrate Specificity , Virus Diseases/genetics
5.
Nucleic Acids Res ; 47(9): 4707-4720, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30916338

ABSTRACT

Members of the ribonuclease (RNase) III family regulate gene expression by processing dsRNAs. It was previously shown that Escherichia coli (Ec) RNase III recognizes dsRNA with little sequence specificity and the cleavage products are mainly 11 nucleotides (nt) long. It was also shown that the mutation of a glutamate (EcE38) to an alanine promotes generation of siRNA-like products typically 22 nt long. To fully characterize substrate specificity and product size of RNase IIIs, we performed in vitro cleavage of dsRNAs by Ec and Aquifex aeolicus (Aa) enzymes and delineated their products by next-generation sequencing. Surprisingly, we found that both enzymes cleave dsRNA at preferred sites, among which a guanine nucleotide was enriched at a specific position (+3G). Based on sequence and structure analyses, we conclude that RNase IIIs recognize +3G via a conserved glutamine (EcQ165/AaQ161) side chain. Abolishing this interaction by mutating the glutamine to an alanine eliminates the observed +3G preference. Furthermore, we identified a second glutamate (EcE65/AaE64), which, when mutated to alanine, also enhances the production of siRNA-like products. Based on these findings, we created a bacterial Dicer that is ideally suited for producing heterogeneous siRNA cocktails to be used in gene silencing studies.


Subject(s)
Mutant Proteins/genetics , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , Ribonuclease III/genetics , Alanine/genetics , Amino Acid Sequence/genetics , Dimerization , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Gene Silencing , Glutamic Acid/genetics , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Mutation , Ribonuclease III/chemistry , Ribonuclease III/isolation & purification , Sequence Analysis, DNA , Substrate Specificity
6.
Nucleic Acids Res ; 47(19): 10134-10150, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31199869

ABSTRACT

Tyrosyl DNA-phosphodiesterase I (TDP1) repairs type IB topoisomerase (TOP1) cleavage complexes generated by TOP1 inhibitors commonly used as anticancer agents. TDP1 also removes DNA 3' end blocking lesions generated by chain-terminating nucleosides and alkylating agents, and base oxidation both in the nuclear and mitochondrial genomes. Combination therapy with TDP1 inhibitors is proposed to synergize with topoisomerase targeting drugs to enhance selectivity against cancer cells exhibiting deficiencies in parallel DNA repair pathways. A crystallographic fragment screening campaign against the catalytic domain of TDP1 was conducted to identify new lead compounds. Crystal structures revealed two fragments that bind to the TDP1 active site and exhibit inhibitory activity against TDP1. These fragments occupy a similar position in the TDP1 active site as seen in prior crystal structures of TDP1 with bound vanadate, a transition state mimic. Using structural insights into fragment binding, several fragment derivatives have been prepared and evaluated in biochemical assays. These results demonstrate that fragment-based methods can be a highly feasible approach toward the discovery of small-molecule chemical scaffolds to target TDP1, and for the first time, we provide co-crystal structures of small molecule inhibitors bound to TDP1, which could serve for the rational development of medicinal TDP1 inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Ligands , Phosphoric Diester Hydrolases/chemistry , Protein Conformation , Base Sequence , Catalytic Domain/genetics , Crystallography , DNA Repair/genetics , Histidine/analogs & derivatives , Histidine/chemistry , Histidine/isolation & purification , Humans , Models, Molecular , Phosphoric Diester Hydrolases/genetics , Signal Transduction , Small Molecule Libraries/chemistry
7.
Angew Chem Int Ed Engl ; 55(19): 5703-7, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27038327

ABSTRACT

Conjugation of the small ubiquitin-like modifier (SUMO) to protein substrates is an important disease-associated posttranslational modification, although few inhibitors of this process are known. Herein, we report the discovery of an allosteric small-molecule binding site on Ubc9, the sole SUMO E2 enzyme. An X-ray crystallographic screen was used to identify two distinct small-molecule fragments that bind to Ubc9 at a site distal to its catalytic cysteine. These fragments and related compounds inhibit SUMO conjugation in biochemical assays with potencies of 1.9-5.8 mm. Mechanistic and biophysical analyses, coupled with molecular dynamics simulations, point toward ligand-induced rigidification of Ubc9 as a mechanism of inhibition.


Subject(s)
Ubiquitin-Conjugating Enzymes/metabolism , Allosteric Regulation , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Ligands , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Substrate Specificity , Sumoylation , Surface Plasmon Resonance , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics
8.
Biochemistry ; 54(11): 2001-10, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25753752

ABSTRACT

The p53 tumor suppressor is a critical mediator of the cellular response to stress. The N-terminal transactivation domain of p53 makes protein interactions that promote its function as a transcription factor. Among those cofactors is the histone acetyltransferase p300, which both stabilizes p53 and promotes local chromatin unwinding. Here, we report the nuclear magnetic resonance solution structure of the Taz2 domain of p300 bound to the second transactivation subdomain of p53. In the complex, p53 forms an α-helix between residues 47 and 55 that interacts with the α1-α2-α3 face of Taz2. Mutational analysis indicated several residues in both p53 and Taz2 that are critical for stabilizing the interaction. Finally, further characterization of the complex by isothermal titration calorimetry revealed that complex formation is pH-dependent and releases a bound chloride ion. This study highlights differences in the structures of complexes formed by the two transactivation subdomains of p53 that may be broadly observed and play critical roles in p53 transcriptional activity.


Subject(s)
E1A-Associated p300 Protein/metabolism , Histone Acetyltransferases/metabolism , Models, Molecular , Tumor Suppressor Protein p53/metabolism , Amino Acid Substitution , Calorimetry, Differential Scanning , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/genetics , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
9.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 7): 1914-21, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25004968

ABSTRACT

Members of the C/EBP family of transcription factors bind to the Taz2 domain of p300/CBP and mediate its phosphorylation through the recruitment of specific kinases. Short sequence motifs termed homology boxes A and B, which comprise their minimal transactivation domains (TADs), are conserved between C/EBP activators and are necessary for specific p300/CBP binding. A possible mode of interaction between C/EBP TADs and the p300 Taz2 domain was implied by the crystal structure of a chimeric protein composed of residues 1723-1818 of p300 Taz2 and residues 37-61 of C/EBPℇ. The segment corresponding to the C/EBPℇ TAD forms two orthogonally disposed helices connected by a short linker and interacts with the core structure of Taz2 from a symmetry-related molecule. It is proposed that other members of the C/EBP family interact with the Taz2 domain in the same manner. The position of the C/EBPℇ peptide on the Taz2 protein interaction surface suggests that the N-termini of C/EBP proteins are unbound in the C/EBP-p300 Taz2 complex. This observation is in agreement with the known location of the docking site of protein kinase HIPK2 in the C/EBPß N-terminus, which associates with the C/EBPß-p300 complex.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , p300-CBP Transcription Factors/chemistry , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/chemistry , Crystallography, X-Ray , Molecular Sequence Data , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid
10.
Proc Natl Acad Sci U S A ; 108(25): 10156-61, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21646538

ABSTRACT

Era, composed of a GTPase domain and a K homology domain, is essential for bacterial cell viability. It is required for the maturation of 16S rRNA and assembly of the 30S ribosomal subunit. We showed previously that the protein recognizes nine nucleotides ( ) near the 3' end of 16S rRNA, and that this recognition stimulates GTP-hydrolyzing activity of Era. In all three kingdoms of life, the sequence and helix 45 (h45) (nucleotides 1506-1529) are highly conserved. It has been shown that the to double mutation severely affects the viability of bacteria. However, whether Era interacts with G1530 and/or h45 and whether such interactions (if any) contribute to the stimulation of Era's GTPase activity were not known. Here, we report two RNA structures that contain nucleotides 1506-1542 (RNA301), one in complex with Era and GDPNP (GNP), a nonhydrolysable GTP-analogue, and the other in complex with Era, GNP, and the KsgA methyltransferase. The structures show that Era recognizes 10 nucleotides, including G1530, and that Era also binds h45. Moreover, GTPase assay experiments show that G1530 does not stimulate Era's GTPase activity. Rather, A1531 and A1534 are most important for stimulation and h45 further contributes to the stimulation. Although G1530 does not contribute to the intrinsic GTPase activity of Era, its interaction with Era is important for binding and is essential for the protein to function, leading to the discovery of a new cold-sensitive phenotype of Era.


Subject(s)
Base Sequence , GTP-Binding Proteins/metabolism , RNA, Ribosomal, 16S/metabolism , RNA-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary/genetics , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , RNA-Binding Proteins/genetics
11.
Anal Biochem ; 436(2): 142-4, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23395976

ABSTRACT

Because of their stringent sequence specificity, the 3C-like proteases from tobacco etch virus (TEV) and human rhinovirus are often used for the removal of affinity tags. The latter enzyme is rumored to have greater catalytic activity at 4 °C, the temperature at which fusion protein substrates are usually digested. Here we report that experiments with fusion protein and peptide substrates confirm this conjecture. Whereas the catalytic efficiency of rhinovirus 3C protease is approximately the same at its optimum temperature (30 °C) and at 4 °C, TEV protease is 10-fold less active at the latter temperature due primarily to a reduction in k(cat).


Subject(s)
Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Rhinovirus/enzymology , Viral Proteins/metabolism , 3C Viral Proteases , Cysteine Endopeptidases/genetics , Endopeptidases/genetics , Kinetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Temperature , Viral Proteins/genetics
12.
Curr Res Struct Biol ; 5: 100095, 2023.
Article in English | MEDLINE | ID: mdl-36820301

ABSTRACT

Dihydroneopterin aldolase (DHNA) is essential for folate biosynthesis in microorganisms. Without a counterpart in mammals, DHNA is an attractive target for antimicrobial agents. Helicobacter pylori infection occurs in human stomach of over 50% of the world population, but first-line therapies for the infection are facing rapidly increasing resistance. Novel antibiotics are urgently needed, toward which structural information on potential targets is critical. We have determined the crystal structure of H. pylori DHNA (HpDHNA) in complex with a pterin molecule (HpDHNA:Pterin) at 1.49-Å resolution. The HpDHNA:Pterin complex forms a tetramer in crystal. The tetramer is also observed in solution by dynamic light scattering and confirmed by small-angle X-ray scattering. To date, all but one reported DHNA structures are octameric complexes. As the only exception, ligand-free Mycobacterium tuberculosis DHNA (apo-MtDHNA) forms a tetramer in crystal, but its active sites are only partially formed. In contrast, the tetrameric HpDHNA:Pterin complex has well-formed active sites. Each active site accommodates one pterin molecule, but the exit of active site is blocked by two amino acid residues exhibiting a contact distance of 5.2 â€‹Å. In contrast, the corresponding contact distance in Staphylococcus aureus DHNA (SaDHNA) is twice the size, ranging from 9.8 to 10.5 â€‹Å, for ligand-free enzyme, the substrate complex, the product complex, and an inhibitor complex. This large contact distance indicates that the active site of SaDHNA is wide open. We propose that this isozyme-specific contact distance (ISCD) is a characteristic feature of DHNA active site. Comparative analysis of HpDHNA and SaDHNA structures suggests a fragment-based strategy for the development of isozyme-specific inhibitors.

13.
RSC Chem Biol ; 4(5): 334-343, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37181631

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a member of the phospholipase D family that can downregulate the anticancer effects of the type I topoisomerase (TOP1) inhibitors by hydrolyzing the 3'-phosphodiester bond between DNA and the TOP1 residue Y723 in the critical stalled intermediate that is the foundation of TOP1 inhibitor mechanism of action. Thus, TDP1 antagonists are attractive as potential enhancers of TOP1 inhibitors. However, the open and extended nature of the TOP1-DNA substrate-binding region has made the development of TDP1 inhibitors extremely challenging. In this study, starting from our recently identified small molecule microarray (SMM)-derived TDP1-inhibitory imidazopyridine motif, we employed a click-based oxime protocol to extend the parent platform into the DNA and TOP1 peptide substrate-binding channels. We applied one-pot Groebke-Blackburn-Bienayme multicomponent reactions (GBBRs) to prepare the needed aminooxy-containing substrates. By reacting these precursors with approximately 250 aldehydes in microtiter format, we screened a library of nearly 500 oximes for their TDP1 inhibitory potencies using an in vitro florescence-based catalytic assay. Select hits were structurally explored as their triazole- and ether-based isosteres. We obtained crystal structures of two of the resulting inhibitors bound to the TDP1 catalytic domain. The structures reveal that the inhibitors form hydrogen bonds with the catalytic His-Lys-Asn triads ("HKN" motifs: H263, K265, N283 and H493, K495, N516), while simultaneously extending into both the substrate DNA and TOP1 peptide-binding grooves. This work provides a structural model for developing multivalent TDP1 inhibitors capable of binding in a tridentate fashion with a central component situated within the catalytic pocket and extensions that project into both the DNA and TOP1 peptide substrate-binding regions.

14.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 3): 201-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22349221

ABSTRACT

The Yersinia pestis YscD protein is an essential component of the type III secretion system. YscD consists of an N-terminal cytoplasmic domain (residues 1-121), a transmembrane linker (122-142) and a large periplasmic domain (143-419). Both the cytoplasmic and the periplasmic domains are required for the assembly of the type III secretion system. Here, the structure of the YscD cytoplasmic domain solved by SAD phasing is presented. Although the three-dimensional structure is similar to those of forkhead-associated (FHA) domains, comparison with the structures of canonical FHA domains revealed that the cytoplasmic domain of YscD lacks the conserved residues that are required for binding phosphothreonine and is therefore unlikely to function as a true FHA domain.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Yersinia pestis/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, Protein , Structural Homology, Protein , Structure-Activity Relationship , Yersinia pestis/metabolism
15.
Proc Natl Acad Sci U S A ; 106(35): 14843-8, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706445

ABSTRACT

ERA, composed of an N-terminal GTPase domain followed by an RNA-binding KH domain, is essential for bacterial cell viability. It binds to 16S rRNA and the 30S ribosomal subunit. However, its RNA-binding site, the functional relationship between the two domains, and its role in ribosome biogenesis remain unclear. We have determined two crystal structures of ERA, a binary complex with GDP and a ternary complex with a GTP-analog and the 1531AUCACCUCCUUA1542 sequence at the 3' end of 16S rRNA. In the ternary complex, the first nine of the 12 nucleotides are recognized by the protein. We show that GTP binding is a prerequisite for RNA recognition by ERA and that RNA recognition stimulates its GTP-hydrolyzing activity. Based on these and other data, we propose a functional cycle of ERA, suggesting that the protein serves as a chaperone for processing and maturation of 16S rRNA and a checkpoint for assembly of the 30S ribosomal subunit. The AUCA sequence is highly conserved among bacteria, archaea, and eukaryotes, whereas the CCUCC, known as the anti-Shine-Dalgarno sequence, is conserved in noneukaryotes only. Therefore, these data suggest a common mechanism for a highly conserved ERA function in all three kingdoms of life by recognizing the AUCA, with a "twist" for noneukaryotic ERA proteins by also recognizing the CCUCC.


Subject(s)
Bacteria/enzymology , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , GTP Phosphohydrolases/genetics , Guanosine Triphosphate/metabolism , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
16.
Front Chem ; 10: 910953, 2022.
Article in English | MEDLINE | ID: mdl-36051621

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (TDP1) repairs stalled type I topoisomerase (TOP1)-DNA complexes by hydrolyzing the phosphodiester bond between the TOP1 Y723 residue and the 3'-phosphate of its DNA substrate. Although TDP1 antagonists could potentially reduce the dose of TOP1 inhibitors needed to achieve effective anticancer effects, the development of validated TDP1 inhibitors has proven to be challenging. This may, in part, be due to the open and extended nature of the TOP1 substrate binding region. We have previously reported imidazopyrazines and imidazopyridines that can inhibit TDP1 catalytic function in vitro. We solved the TDP1 crystal structures with bound inhibitors of this class and found that the dicarboxylic acid functionality within the N-(3,4-dicarboxyphenyl)-2-diphenylimidazo [1,2-a]pyridin-3-amine platform overlaps with aspects of phosphoryl substrate recognition. Yet phosphonic acids could potentially better-replicate cognate TOP1-DNA substrate binding interactions than carboxylic acids. As reported herein, we designed phosphonic acid-containing variants of our previously reported carboxylic acid-containing imidazopyrazine and imidazopyridine inhibitors and effected their synthesis using one-pot Groebke-Blackburn-Bienayme multicomponent reactions. We obtained crystal structures of TDP1 complexed with a subset of inhibitors. We discuss binding interactions of these inhibitors within the context of phosphate-containing substrate and carboxylic acid-based inhibitors. These compounds represent a new structural class of small molecule ligands that mimic aspects of the 3'-processed substrate that results from TDP1 catalysis.

17.
J Struct Biol ; 176(3): 292-301, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21963792

ABSTRACT

Chk2 (checkpoint kinase 2) is a serine/threonine kinase that participates in a series of signaling networks responsible for maintaining genomic integrity and responding to DNA damage. The development of selective Chk2 inhibitors has recently attracted much interest as a means of sensitizing cancer cells to current DNA-damaging agents used in the treatment of cancer. Additionally, selective Chk2 inhibitors may reduce p53-mediated apoptosis in normal tissues, thereby helping to mitigate adverse side effects from chemotherapy and radiation. Thus far, relatively few selective inhibitors of Chk2 have been described and none have yet progressed into clinical trials. Here, we report crystal structures of the catalytic domain of Chk2 in complex with a novel series of potent and selective small molecule inhibitors. These compounds exhibit nanomolar potencies and are selective for Chk2 over Chk1. The structures reported here elucidate the binding modes of these inhibitors to Chk2 and provide information that can be exploited for the structure-assisted design of novel chemotherapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Catalytic Domain , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Binding Sites , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Crystallography, X-Ray , Humans , Molecular Structure , Molecular Targeted Therapy , Protein Binding , Protein Kinases/chemistry
18.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 5): 471-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21543850

ABSTRACT

There are over 100 genes in the human genome that encode protein tyrosine phosphatases (PTPs) and approximately 60 of these are classified as dual-specificity phosphatases (DUSPs). Although many dual-specificity phosphatases are still not well characterized, novel functions have been discovered for some of them that have led to new insights into a variety of biological processes and the molecular basis for certain diseases. Indeed, as the functions of DUSPs continue to be elucidated, a growing number of them are emerging as potential therapeutic targets for diseases such as cancer, diabetes and inflammatory disorders. Here, the overexpression, purification and structure determination of DUSP27 at 2.38 Šresolution are presented.


Subject(s)
Phosphoprotein Phosphatases/chemistry , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Humans , Models, Molecular , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/isolation & purification , Structural Homology, Protein , Up-Regulation
19.
Protein Expr Purif ; 77(1): 53-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21073956

ABSTRACT

Carboxypeptidases may serve as tools for removal of C-terminal affinity tags. In the present study, we describe the expression and purification of an A-type carboxypeptidase from the fungal pathogen Metarhizium anisopliae (MeCPA) that has been genetically engineered to facilitate the removal of polyhistidine tags from the C-termini of recombinant proteins. A complete, systematic analysis of the specificity of MeCPA in comparison with that of bovine carboxypeptidase A (BoCPA) was carried out. Our results indicate that the specificity of the two enzymes is similar but not identical. Histidine residues are removed more efficiently by MeCPA. The very inefficient digestion of peptides with C-terminal lysine or arginine residues, along with the complete inability of the enzyme to remove a C-terminal proline, suggests a strategy for designing C-terminal affinity tags that can be trimmed by MeCPA (or BoCPA) to produce a digestion product with a homogeneous endpoint.


Subject(s)
Affinity Labels/metabolism , Carboxypeptidases A/metabolism , Cattle/metabolism , Histidine/metabolism , Metarhizium/enzymology , Affinity Labels/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Carboxypeptidases A/chemistry , Histidine/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sodium Chloride , Substrate Specificity
20.
Structure ; 17(3): 374-85, 2009 Mar 11.
Article in English | MEDLINE | ID: mdl-19278652

ABSTRACT

Among methyltransferases, KsgA and the reaction it catalyzes are conserved throughout evolution. However, the specifics of substrate recognition by the enzyme remain unknown. Here we report structures of Aquifex aeolicus KsgA, in its ligand-free form, in complex with RNA, and in complex with both RNA and S-adenosylhomocysteine (SAH, reaction product of cofactor S-adenosylmethionine), revealing critical structural information on KsgA-RNA and KsgA-SAH interactions. Moreover, the structures show how conformational changes that occur upon RNA binding create the cofactor-binding site. There are nine conserved functional motifs (motifs I-VIII and X) in KsgA. Prior to RNA binding, motifs I and VIII are flexible, each exhibiting two distinct conformations. Upon RNA binding, the two motifs become stabilized in one of these conformations, which is compatible with the binding of SAH. Motif X, which is also stabilized upon RNA binding, is directly involved in the binding of SAH.


Subject(s)
Coenzymes/chemistry , Methyltransferases/chemistry , RNA/chemistry , S-Adenosylhomocysteine/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , Catalysis , Coenzymes/metabolism , Crystallography, X-Ray , Gram-Negative Bacteria/enzymology , Ligands , Methyltransferases/metabolism , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Conformation , RNA/metabolism , S-Adenosylhomocysteine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL