Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Med Res Rev ; 39(2): 684-705, 2019 03.
Article in English | MEDLINE | ID: mdl-30192413

ABSTRACT

Escherichia coli Dihydrofolate reductase is an important enzyme that is essential for the survival of the Gram-negative microorganism. Inhibitors designed against this enzyme have demonstrated application as antibiotics. However, either because of poor bioavailability of the small-molecules resulting from their inability to cross the double membrane in Gram-negative bacteria or because the microorganism develops resistance to the antibiotics by mutating the DHFR target, discovery of new antibiotics against the enzyme is mandatory to overcome drug-resistance. This review summarizes the field of DHFR inhibition with special focus on recent efforts to effectively interface computational and experimental efforts to discover novel classes of inhibitors that target allosteric and active-sites in drug-resistant variants of EcDHFR.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/chemistry , Algorithms , Allosteric Site , Animals , Catalytic Domain , Drug Design , Drug Discovery , Humans , Ligands , Permeability/drug effects , Structure-Activity Relationship
2.
Bioinformatics ; 31(4): 539-44, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25336501

ABSTRACT

MOTIVATION: Shape-based alignment of small molecules is a widely used approach in computer-aided drug discovery. Most shape-based ligand structure alignment applications, both commercial and freely available ones, use the Tanimoto coefficient or similar functions for evaluating molecular similarity. Major drawbacks of using such functions are the size dependence of the score and the fact that the statistical significance of the molecular match using such metrics is not reported. RESULTS: We describe a new open-source ligand structure alignment and virtual screening (VS) algorithm, LIGSIFT, that uses Gaussian molecular shape overlay for fast small molecule alignment and a size-independent scoring function for efficient VS based on the statistical significance of the score. LIGSIFT was tested against the compounds for 40 protein targets available in the Directory of Useful Decoys and the performance was evaluated using the area under the ROC curve (AUC), the Enrichment Factor (EF) and Hit Rate (HR). LIGSIFT-based VS shows an average AUC of 0.79, average EF values of 20.8 and a HR of 59% in the top 1% of the screened library. AVAILABILITY AND IMPLEMENTATION: LIGSIFT software, including the source code, is freely available to academic users at http://cssb.biology.gatech.edu/LIGSIFT. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online. CONTACT: skolnick@gatech.edu.


Subject(s)
Algorithms , Databases, Chemical , Drug Discovery/methods , Molecular Docking Simulation , Proteins/chemistry , Software , Binding Sites , Drug Evaluation, Preclinical , Humans , Ligands , Models, Molecular , Protein Conformation , Proteins/metabolism , User-Computer Interface
3.
Bioorg Med Chem Lett ; 25(6): 1163-70, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25690787

ABSTRACT

Coincidence of the properties of ligand binding pockets in native proteins with those in proteins generated by computer simulations without selection for function shows that pockets are a generic protein feature and the number of distinct pockets is small. Similar pockets occur in unrelated protein structures, an observation successfully employed in pocket-based virtual ligand screening. The small number of pockets suggests that off-target interactions among diverse proteins are inherent; kinases, proteases and phosphatases show this prototypical behavior. The ability to repurpose FDA approved drugs is general, and minor side effects cannot be avoided. Finally, the implications to drug discovery are explored.


Subject(s)
Evolution, Molecular , Ligands , Proteins/chemistry , Binding Sites , Drug Evaluation, Preclinical , Molecular Dynamics Simulation , Protein Binding , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Structure, Tertiary , Proteins/metabolism
4.
J Chem Inf Model ; 55(8): 1757-70, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26225536

ABSTRACT

Often in pharmaceutical research the goal is to identify small molecules that can interact with and appropriately modify the biological behavior of a new protein target. Unfortunately, most proteins lack both known structures and small molecule binders, prerequisites of many virtual screening, VS, approaches. For such proteins, ligand homology modeling, LHM, that copies ligands from homologous and perhaps evolutionarily distant template proteins, has been shown to be a powerful VS approach to identify possible binding ligands. However, if we want to target a specific pocket for which there is no homologous holo template protein structure, then LHM will not work. To address this issue, in a new pocket-based approach, PoLi, we generalize LHM by exploiting the fact that the number of distinct small molecule ligand-binding pockets in proteins is small. PoLi identifies similar ligand-binding pockets in a holo template protein library, selectively copies relevant parts of template ligands, and uses them for VS. In practice, PoLi is a hybrid structure and ligand-based VS algorithm that integrates 2D fingerprint-based and 3D shape-based similarity metrics for improved virtual screening performance. On standard DUD and DUD-E benchmark databases, using modeled receptor structures, PoLi achieves an average enrichment factor of 13.4 and 9.6, respectively, in the top 1% of the screened library. In contrast, traditional docking-based VS using AutoDock Vina and homology-based VS using FINDSITE(filt) have an average enrichment of 1.6 (3.0) and 9.0 (7.9) on the DUD (DUD-E) sets, respectively. Experimental validation of PoLi predictions on dihydrofolate reductase, DHFR, using differential scanning fluorimetry, DSF, identifies multiple ligands with diverse molecular scaffolds, thus demonstrating the advantage of PoLi over current state-of-the-art VS methods.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Algorithms , Binding Sites , Databases, Protein , Drug Discovery , Ligands , Molecular Docking Simulation , Protein Conformation
5.
Nucleic Acids Res ; 41(Database issue): D1096-103, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23087378

ABSTRACT

BioLiP (http://zhanglab.ccmb.med.umich.edu/BioLiP/) is a semi-manually curated database for biologically relevant ligand-protein interactions. Establishing interactions between protein and biologically relevant ligands is an important step toward understanding the protein functions. Most ligand-binding sites prediction methods use the protein structures from the Protein Data Bank (PDB) as templates. However, not all ligands present in the PDB are biologically relevant, as small molecules are often used as additives for solving the protein structures. To facilitate template-based ligand-protein docking, virtual ligand screening and protein function annotations, we develop a hierarchical procedure for assessing the biological relevance of ligands present in the PDB structures, which involves a four-step biological feature filtering followed by careful manual verifications. This procedure is used for BioLiP construction. Each entry in BioLiP contains annotations on: ligand-binding residues, ligand-binding affinity, catalytic sites, Enzyme Commission numbers, Gene Ontology terms and cross-links to the other databases. In addition, to facilitate the use of BioLiP for function annotation of uncharacterized proteins, a new consensus-based algorithm COACH is developed to predict ligand-binding sites from protein sequence or using 3D structure. The BioLiP database is updated weekly and the current release contains 204 223 entries.


Subject(s)
Databases, Protein , Ligands , Proteins/chemistry , Algorithms , Binding Sites , Internet , Proteins/metabolism
6.
Bioinformatics ; 29(20): 2588-95, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23975762

ABSTRACT

MOTIVATION: Identification of protein-ligand binding sites is critical to protein function annotation and drug discovery. However, there is no method that could generate optimal binding site prediction for different protein types. Combination of complementary predictions is probably the most reliable solution to the problem. RESULTS: We develop two new methods, one based on binding-specific substructure comparison (TM-SITE) and another on sequence profile alignment (S-SITE), for complementary binding site predictions. The methods are tested on a set of 500 non-redundant proteins harboring 814 natural, drug-like and metal ion molecules. Starting from low-resolution protein structure predictions, the methods successfully recognize >51% of binding residues with average Matthews correlation coefficient (MCC) significantly higher (with P-value <10(-9) in student t-test) than other state-of-the-art methods, including COFACTOR, FINDSITE and ConCavity. When combining TM-SITE and S-SITE with other structure-based programs, a consensus approach (COACH) can increase MCC by 15% over the best individual predictions. COACH was examined in the recent community-wide COMEO experiment and consistently ranked as the best method in last 22 individual datasets with the Area Under the Curve score 22.5% higher than the second best method. These data demonstrate a new robust approach to protein-ligand binding site recognition, which is ready for genome-wide structure-based function annotations. AVAILABILITY: http://zhanglab.ccmb.med.umich.edu/COACH/


Subject(s)
Proteins/chemistry , Sequence Alignment/methods , Sequence Analysis, Protein/methods , Algorithms , Binding Sites , Databases, Protein , Humans , Ligands , Models, Molecular , Protein Structure, Tertiary , Proteins/metabolism
7.
Nucleic Acids Res ; 40(Web Server issue): W471-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22570420

ABSTRACT

We have developed a new COFACTOR webserver for automated structure-based protein function annotation. Starting from a structural model, given by either experimental determination or computational modeling, COFACTOR first identifies template proteins of similar folds and functional sites by threading the target structure through three representative template libraries that have known protein-ligand binding interactions, Enzyme Commission number or Gene Ontology terms. The biological function insights in these three aspects are then deduced from the functional templates, the confidence of which is evaluated by a scoring function that combines both global and local structural similarities. The algorithm has been extensively benchmarked by large-scale benchmarking tests and demonstrated significant advantages compared to traditional sequence-based methods. In the recent community-wide CASP9 experiment, COFACTOR was ranked as the best method for protein-ligand binding site predictions. The COFACTOR sever and the template libraries are freely available at http://zhanglab.ccmb.med.umich.edu/COFACTOR.


Subject(s)
Algorithms , Molecular Sequence Annotation , Proteins/chemistry , Proteins/physiology , Software , Binding Sites , Enzymes/chemistry , Enzymes/physiology , Internet , Ligands , Protein Conformation , Proteins/genetics
9.
J Proteome Res ; 10(12): 5503-11, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22003824

ABSTRACT

Alternative splicing allows a single gene to generate multiple mRNA transcripts, which can be translated into functionally diverse proteins. However, experimentally determined structures of protein splice isoforms are rare, and homology modeling methods are poor at predicting atomic-level structural differences because of high sequence identity. Here we exploit the state-of-the-art structure prediction method I-TASSER to analyze the structural and functional consequences of alternative splicing of proteins differentially expressed in a breast cancer model. We first successfully benchmarked the I-TASSER pipeline for structure modeling of all seven pairs of protein splice isoforms, which are known to have experimentally solved structures. We then modeled three cancer-related variant pairs reported to have opposite functions. In each pair, we observed structural differences in regions where the presence or absence of a motif can directly influence the distinctive functions of the variants. Finally, we applied the method to five splice variants overexpressed in mouse Her2/neu mammary tumor: anxa6, calu, cdc42, ptbp1, and tax1bp3. Despite >75% sequence identity between the variants, structural differences were observed in biologically important regions of these protein pairs. These results demonstrate the feasibility of integrating proteomic analysis with structure-based conformational predictions of differentially expressed alternative splice variants in cancers and other conditions.


Subject(s)
Alternative Splicing , Genes, erbB-2 , Mammary Neoplasms, Experimental/metabolism , Proteomics/methods , Algorithms , Amino Acid Sequence , Animals , Annexin A6/genetics , Annexin A6/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Computational Biology/methods , Databases, Protein , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Models, Molecular , Molecular Sequence Data , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Protein Conformation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Structure-Activity Relationship , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
10.
Proteins ; 79 Suppl 10: 147-60, 2011.
Article in English | MEDLINE | ID: mdl-22069036

ABSTRACT

I-TASSER is an automated pipeline for protein tertiary structure prediction using multiple threading alignments and iterative structure assembly simulations. In CASP9 experiments, two new algorithms, QUARK and fragment-guided molecular dynamics (FG-MD), were added to the I-TASSER pipeline for improving the structural modeling accuracy. QUARK is a de novo structure prediction algorithm used for structure modeling of proteins that lack detectable template structures. For distantly homologous targets, QUARK models are found useful as a reference structure for selecting good threading alignments and guiding the I-TASSER structure assembly simulations. FG-MD is an atomic-level structural refinement program that uses structural fragments collected from the PDB structures to guide molecular dynamics simulation and improve the local structure of predicted model, including hydrogen-bonding networks, torsion angles, and steric clashes. Despite considerable progress in both the template-based and template-free structure modeling, significant improvements on protein target classification, domain parsing, model selection, and ab initio folding of ß-proteins are still needed to further improve the I-TASSER pipeline.


Subject(s)
Computational Biology/methods , Molecular Dynamics Simulation , Proteins/chemistry , Algorithms , Databases, Protein , Protein Folding , Protein Structure, Tertiary , Software
11.
Genomics Proteomics Bioinformatics ; 19(6): 998-1011, 2021 12.
Article in English | MEDLINE | ID: mdl-33631427

ABSTRACT

The number of available protein sequences in public databases is increasing exponentially. However, a significant percentage of these sequences lack functional annotation, which is essential for the understanding of how biological systems operate. Here, we propose a novel method, Quantitative Annotation of Unknown STructure (QAUST), to infer protein functions, specifically Gene Ontology (GO) terms and Enzyme Commission (EC) numbers. QAUST uses three sources of information: structure information encoded by global and local structure similarity search, biological network information inferred by protein-protein interaction data, and sequence information extracted from functionally discriminative sequence motifs. These three pieces of information are combined by consensus averaging to make the final prediction. Our approach has been tested on 500 protein targets from the Critical Assessment of Functional Annotation (CAFA) benchmark set. The results show that our method provides accurate functional annotation and outperforms other prediction methods based on sequence similarity search or threading. We further demonstrate that a previously unknown function of human tripartite motif-containing 22 (TRIM22) protein predicted by QAUST can be experimentally validated.


Subject(s)
Proteins , Software , Computational Biology/methods , Databases, Protein , Humans , Proteins/chemistry , Proteins/genetics
12.
Nat Commun ; 9(1): 219, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335539

ABSTRACT

Neurological drugs are often associated with serious side effects, yet drug screens typically focus only on efficacy. We demonstrate a novel paradigm utilizing high-throughput in vivo electrophysiology and brain activity patterns (BAPs). A platform with high sensitivity records local field potentials (LFPs) simultaneously from many zebrafish larvae over extended periods. We show that BAPs from larvae experiencing epileptic seizures or drug-induced side effects have substantially reduced complexity (entropy), similar to reduced LFP complexity observed in Parkinson's disease. To determine whether drugs that enhance BAP complexity produces positive outcomes, we used light pulses to trigger seizures in a model of Dravet syndrome, an intractable genetic epilepsy. The highest-ranked compounds identified by BAP analysis exhibit far greater anti-seizure efficacy and fewer side effects during subsequent in-depth behavioral assessment. This high correlation with behavioral outcomes illustrates the power of brain activity pattern-based screens and identifies novel therapeutic candidates with minimal side effects.


Subject(s)
Brain/physiopathology , Electrophysiological Phenomena , Psychotropic Drugs/pharmacology , Zebrafish/physiology , Animals , Disease Models, Animal , Electrophysiology/methods , Epilepsies, Myoclonic/diagnosis , Epilepsies, Myoclonic/physiopathology , Humans , Larva/drug effects , Larva/genetics , Larva/physiology , Psychotropic Drugs/toxicity , Zebrafish/genetics
13.
Drug Discov Today ; 19(9): 1353-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24980786

ABSTRACT

The Computational Analysis of Novel Drug Opportunities (CANDO) platform (http://protinfo.org/cando) uses similarity of compound-proteome interaction signatures to infer homology of compound/drug behavior. We constructed interaction signatures for 3733 human ingestible compounds covering 48,278 protein structures mapping to 2030 indications based on basic science methodologies to predict and analyze protein structure, function, and interactions developed by us and others. Our signature comparison and ranking approach yielded benchmarking accuracies of 12-25% for 1439 indications with at least two approved compounds. We prospectively validated 49/82 'high value' predictions from nine studies covering seven indications, with comparable or better activity to existing drugs, which serve as novel repurposed therapeutics. Our approach may be generalized to compounds beyond those approved by the FDA, and can also consider mutations in protein structures to enable personalization. Our platform provides a holistic multiscale modeling framework of complex atomic, molecular, and physiological systems with broader applications in medicine and engineering.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Proteins/metabolism , Animals , Drug Design , Drug Repositioning , Humans , Models, Theoretical , Protein Conformation , Proteins/chemistry , Proteome
14.
Structure ; 20(6): 987-97, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22560732

ABSTRACT

Proteins perform functions through interacting with other molecules. However, structural details for most of the protein-ligand interactions are unknown. We present a comparative approach (COFACTOR) to recognize functional sites of protein-ligand interactions using low-resolution protein structural models, based on a global-to-local sequence and structural comparison algorithm. COFACTOR was tested on 501 proteins, which harbor 582 natural and drug-like ligand molecules. Starting from I-TASSER structure predictions, the method successfully identifies ligand-binding pocket locations for 65% of apo receptors with an average distance error 2 Å. The average precision of binding-residue assignments is 46% and 137% higher than that by FINDSITE and ConCavity. In CASP9, COFACTOR achieved a binding-site prediction precision 72% and Matthews correlation coefficient 0.69 for 31 blind test proteins, which was significantly higher than all other participating methods. These data demonstrate the power of structure-based approaches to protein-ligand interaction predictions applicable for genome-wide structural and functional annotations.


Subject(s)
Algorithms , Models, Molecular , Proteins/chemistry , Software , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Computer Simulation , Ligands , Protein Binding , Protein Structure, Tertiary , Structural Homology, Protein
15.
Open Biochem J ; 5: 9-26, 2011.
Article in English | MEDLINE | ID: mdl-21633666

ABSTRACT

Parkin belongs to a class of multiple RING domain proteins designated as RBR (RING, in between RING, RING) proteins. In this review we examine what is known regarding the structure/function relationship of the Parkin protein. Parkin contains three RING domains plus a ubiquitin-like domain and an in-between-RING (IBR) domain. RING domains are rich in cysteine amino acids that act as ligands to bind zinc ions. RING domains may interact with DNA or with other proteins and perform a wide range of functions. Some function as E3 ubiquitin ligases, participating in attachment of ubiquitin chains to signal proteasome degradation; however, ubiquitin may be attached for purposes other than proteasome degradation. It was determined that the C-terminal most RING, RING2, is essential for Parkin to function as an E3 ubiquitin ligase and a number of substrates have been identified. However, Parkin also participates in a number of other fiunctions, such as DNA repair, microtubule stabilization, and formation of aggresomes. Some functions, such as participation in a multi-protein complex implicated in NMDA activity at the post synaptic density, do not require ubiquitination of substrate molecules. Recent observations of RING proteins suggest their function may be regulated by zinc ion binding. We have modeled the three RING domains of Parkin and have identified a new set of RING2 ligands. This set allows for binding of two rather than just one zinc ion, opening the possibility that the number of zinc ions bound acts as a molecular switch to modulate Parkin function.

16.
J Vis Exp ; (57): e3259, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-22082966

ABSTRACT

Genome sequencing projects have ciphered millions of protein sequence, which require knowledge of their structure and function to improve the understanding of their biological role. Although experimental methods can provide detailed information for a small fraction of these proteins, computational modeling is needed for the majority of protein molecules which are experimentally uncharacterized. The I-TASSER server is an on-line workbench for high-resolution modeling of protein structure and function. Given a protein sequence, a typical output from the I-TASSER server includes secondary structure prediction, predicted solvent accessibility of each residue, homologous template proteins detected by threading and structure alignments, up to five full-length tertiary structural models, and structure-based functional annotations for enzyme classification, Gene Ontology terms and protein-ligand binding sites. All the predictions are tagged with a confidence score which tells how accurate the predictions are without knowing the experimental data. To facilitate the special requests of end users, the server provides channels to accept user-specified inter-residue distance and contact maps to interactively change the I-TASSER modeling; it also allows users to specify any proteins as template, or to exclude any template proteins during the structure assembly simulations. The structural information could be collected by the users based on experimental evidences or biological insights with the purpose of improving the quality of I-TASSER predictions. The server was evaluated as the best programs for protein structure and function predictions in the recent community-wide CASP experiments. There are currently >20,000 registered scientists from over 100 countries who are using the on-line I-TASSER server.


Subject(s)
Algorithms , Proteins/chemistry , Proteins/physiology , Software , Computer Simulation , Databases, Protein , Models, Chemical , Models, Molecular , Protein Conformation , Structure-Activity Relationship
17.
Nat Protoc ; 5(4): 725-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20360767

ABSTRACT

The iterative threading assembly refinement (I-TASSER) server is an integrated platform for automated protein structure and function prediction based on the sequence-to-structure-to-function paradigm. Starting from an amino acid sequence, I-TASSER first generates three-dimensional (3D) atomic models from multiple threading alignments and iterative structural assembly simulations. The function of the protein is then inferred by structurally matching the 3D models with other known proteins. The output from a typical server run contains full-length secondary and tertiary structure predictions, and functional annotations on ligand-binding sites, Enzyme Commission numbers and Gene Ontology terms. An estimate of accuracy of the predictions is provided based on the confidence score of the modeling. This protocol provides new insights and guidelines for designing of online server systems for the state-of-the-art protein structure and function predictions. The server is available at http://zhanglab.ccmb.med.umich.edu/I-TASSER.


Subject(s)
Databases, Protein , Internet , Proteins/chemistry , Proteins/genetics , Sequence Alignment , Structural Homology, Protein , Amino Acid Sequence , Computer Simulation , Databases, Protein/statistics & numerical data , Models, Molecular , Online Systems , Proteins/physiology , Sequence Alignment/statistics & numerical data
18.
PLoS One ; 4(8): e6701, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19693270

ABSTRACT

Hydrogen constitutes nearly half of all atoms in proteins and their positions are essential for analyzing hydrogen-bonding interactions and refining atomic-level structures. However, most protein structures determined by experiments or computer prediction lack hydrogen coordinates. We present a new algorithm, HAAD, to predict the positions of hydrogen atoms based on the positions of heavy atoms. The algorithm is built on the basic rules of orbital hybridization followed by the optimization of steric repulsion and electrostatic interactions. We tested the algorithm using three independent data sets: ultra-high-resolution X-ray structures, structures determined by neutron diffraction, and NOE proton-proton distances. Compared with the widely used programs CHARMM and REDUCE, HAAD has a significantly higher accuracy, with the average RMSD of the predicted hydrogen atoms to the X-ray and neutron diffraction structures decreased by 26% and 11%, respectively. Furthermore, hydrogen atoms placed by HAAD have more matches with the NOE restraints and fewer clashes with heavy atoms. The average CPU cost by HAAD is 18 and 8 times lower than that of CHARMM and REDUCE, respectively. The significant advantage of HAAD in both the accuracy and the speed of the hydrogen additions should make HAAD a useful tool for the detailed study of protein structure and function. Both an executable and the source code of HAAD are freely available at http://zhang.bioinformatics.ku.edu/HAAD.


Subject(s)
Algorithms , Hydrogen/chemistry , Protein Conformation , Proteins/chemistry , Magnetic Resonance Spectroscopy , X-Ray Diffraction
19.
In Silico Biol ; 9(1-2): S41-55, 2009.
Article in English | MEDLINE | ID: mdl-19537164

ABSTRACT

Using a large database of protein domain families of known 3-D structure we present an analysis on the relationships among sequences, structures and functions of closely-related enzymes performed at the level of catalytic domains. Only in 38% of the pairs of homologous catalytic domains characterized by over about 60% of sequence identity the functions are almost completely identical. Nearly 43% of the pairs differ in their substrate specificity. Hence the most common variation of enzyme function among the closely-related homologues is the differences in the substrate specificity. For homologous pairs characterized by a sequence identity of 30-60%, if the structural difference metric is less than about 30, the functions are highly conserved. For clearly homologous protein domain pairs, usually sharing less than 40% sequence identity, we observe that often the chemical groups involved in the functions, and the cofactors differ. We also report of extremely unusual cases of closely-related homologues belonging to entirely different classes of enzymes. Such drastic shifts in the gross functions of homologues seem to be achieved by retooling of catalytic residues or by altering the stability of the intermediates in the biochemical reactions. Our work provides guidelines on the functional annotation based on homology searches and in structural genomics initiatives.


Subject(s)
Computational Biology , Enzymes/chemistry , Enzymes/metabolism , Evolution, Molecular , Genomics , Enzymes/classification , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment
20.
J Pharm Sci ; 98(10): 3562-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19199298

ABSTRACT

The objective of this work was to evaluate the solution stability of the EC1 domain of E-cadherin under various conditions. The EC1 domain was incubated at various temperatures (4, 37, and 70 degrees C) and pH values (3.0, 7.0, and 9.0). At pH 9.0 and 37 or 70 degrees C, a significant loss of EC1 was observed due to precipitation and a hydrolysis reaction. The degradation was suppressed upon addition of dithiothreitol (DTT), suggesting that the formation of EC1 dimer facilitated the EC1 degradation. At 4 degrees C and various pH values, the EC1 secondary and tertiary showed changes upon incubation up to 28 days, and DTT prevented any structural changes upon 28 days of incubation. Molecular dynamics simulations indicated that the dimer of EC1 has higher mobility than does the monomer; this higher mobility of the EC1 dimer may contribute to instability of the EC1 domain.


Subject(s)
Cadherins/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Circular Dichroism , Computer Simulation , Dimerization , Dithiothreitol/analysis , Drug Stability , Escherichia coli/genetics , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Fluorescence , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL