Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791574

ABSTRACT

Being a component of the Ras/Raf/MEK/ERK signaling pathway crucial for cellular responses, the VRAF murine sarcoma viral oncogene homologue B1 (BRAF) kinase has emerged as a promising target for anticancer drug discovery due to oncogenic mutations that lead to pathway hyperactivation. Despite the discovery of several small-molecule BRAF kinase inhibitors targeting oncogenic mutants, their clinical utility has been limited by challenges such as off-target effects and suboptimal pharmacological properties. This study focuses on identifying miniprotein inhibitors for the oncogenic V600E mutant BRAF, leveraging their potential as versatile drug candidates. Using a structure-based de novo design approach based on binding affinity to V600E mutant BRAF and hydration energy, 39 candidate miniprotein inhibitors comprising three helices and 69 amino acids were generated from the substructure of the endogenous ligand protein (14-3-3). Through in vitro binding and kinase inhibition assays, two miniproteins (63 and 76) were discovered as novel inhibitors of V600E mutant BRAF with low-micromolar activity, with miniprotein 76 demonstrating a specific impediment to MEK1 phosphorylation in mammalian cells. These findings highlight miniprotein 76 as a potential lead compound for developing new cancer therapeutics, and the structural features contributing to its biochemical potency against V600E mutant BRAF are discussed in detail.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Mutation , Drug Discovery/methods , Phosphorylation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Design , Protein Binding , Structure-Activity Relationship , Models, Molecular
2.
STAR Protoc ; 5(2): 102999, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38573865

ABSTRACT

The microbial transcription factor YhaJ responds to 2,4-dinitrotoluene (DNT) derivatives. Here, we describe steps for overexpression and purification of the protein, characterization for the binding of a DNT derivative methylhydroquinone, and crystallization by using a random seeding technique. We then detail procedures for structure determination by employing the crystal-twin resolving processes. This protocol can also be performed using other DNT derivatives. For complete details on the use and execution of this protocol, please refer to Kim et al.1.


Subject(s)
Crystallization , Dinitrobenzenes/chemistry , Crystallography, X-Ray/methods , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism
3.
iScience ; 26(10): 107984, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37822509

ABSTRACT

Detection of landmines without harming personnel is a global issue. The bacterial transcription factor YhaJ selectively detects metabolites of explosives, and it can be used as a key component of DNT biosensors. However, the wild-type YhaJ has a binding affinity that is not sufficient for the detection of trace amounts of explosives leaked from landmines buried in the soil. Here, we report crystal structures of the effector-binding domain of YhaJ in both the apo- and effector-bound forms. A structural comparison of the two forms revealed that the loop above the primary effector-binding site significantly switches its conformation upon effector binding. The primary effector-binding site involves hydrophobic and polar interactions, having specificity to hydroxyl-substituted benzene compounds. The structures explain the mechanism of activity-enhancing mutations and provide information for the rational engineering of YhaJ biosensors for the sensitive detection of explosives.

4.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 7): 265-269, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35787553

ABSTRACT

Receptor-type protein tyrosine phosphatases (RPTPs) receive extracellular stimuli and transfer them into cells. They regulate cell growth, differentiation and death via specific signals. They have also been implicated in cancer, diabetes and neurological diseases. RPTPH, a member of the type 3 RPTP (R3-PTP) family, is an important regulator of colorectal cancer and hepatic carcinoma. Despite its importance in drug development, the structure of RPTPH has not yet been resolved. Here, the crystal structure of the catalytic domain of RPTPH was determined at 1.56 Šresolution. Despite similarities to other R3-PTPs in its overall structure, RPTPH exhibited differences in its loop regions and side-chain conformations. Compared with other R3-PTPs, RPTPH has unique side chains near its active site that may confer specificity for inhibitor binding. Therefore, detailed information on the structure of RPTPH provides clues for the development of specific inhibitors.


Subject(s)
Protein Tyrosine Phosphatases , Catalytic Domain , Crystallography, X-Ray , Humans , Protein Tyrosine Phosphatases/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
5.
Article in English | MEDLINE | ID: mdl-34913730

ABSTRACT

Aims: Mitochondrial respiratory supercomplexes mediate redox electron transfer, generating a proton gradient for ATP synthesis. To provide structural information on the function of supercomplexes in physiologically relevant conditions, we conducted cryoelectron microscopy studies with supercomplexes in a lipid-preserving state. Results: Here, we present cryoelectron microscopy structures of bovine respiratory supercomplex I1III2IV1 by using a lipid-preserving sample preparation. The preparation greatly enhances the intercomplex quinone transfer activity. The structures reveal large intercomplex motions that result in different shapes and sizes of the intercomplex space between complexes I and III, forming a dynamic substrate pool. Biochemical and structural analyses indicated that intercomplex phospholipids mediate the intercomplex motions. An analysis of the different classes of focus-refined complex I showed that structural switches due to quinone reduction led to the formation of a novel channel that could transfer reduced quinones to the intercomplex substrate pool. Innovation and Conclusion: Our results indicate potential mechanism for the facilitated electron transfer involving a dynamic substrate pool and intercomplex movement by which supercomplexes play an active role in the regulation of metabolic flux and reactive oxygen species.

6.
Sci Rep ; 11(1): 16765, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34408201

ABSTRACT

When primary cancer faces limited oxygen and nutrient supply, it undergoes an epithelial-mesenchymal transition, which increases cancer cell motility and invasiveness. The migratory and invasive cancer cells often exert aggressive cancer development or even cancer metastasis. In this study, we investigated a novel compound, 3-acetyl-5,8-dichloro-2-((2,4-dichlorophenyl)amino)quinolin-4(1H)-one (ADQ), that showed significant suppression of wound healing and cellular invasion. This compound also inhibited anchorage-independent cell growth, multicellular tumor spheroid survival/invasion, and metalloprotease activities. The anti-proliferative effects of ADQ were mediated by inhibition of the Akt pathway. In addition, ADQ reduced the expression of mesenchymal markers of cancer cells, which was associated with the suppressed expression of Twist1. In conclusion, ADQ successfully suppressed carcinogenic activity by inhibiting the Akt signaling pathway and Twist1, which suggests that ADQ may be an efficient candidate for cancer drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Liver Neoplasms , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Twist-Related Protein 1/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplasm Invasiveness , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Twist-Related Protein 1/genetics
7.
EMBO Rep ; 22(6): e51323, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33938112

ABSTRACT

In eukaryotic cells, mitochondria are closely tethered to the endoplasmic reticulum (ER) at sites called mitochondria-associated ER membranes (MAMs). Ca2+ ion and phospholipid transfer occurs at MAMs to support diverse cellular functions. Unlike those in yeast, the protein complexes involved in phospholipid transfer at MAMs in humans have not been identified. Here, we determine the crystal structure of the tetratricopeptide repeat domain of PTPIP51 (PTPIP51_TPR), a mitochondrial protein that interacts with the ER-anchored VAPB protein at MAMs. The structure of PTPIP51_TPR shows an archetypal TPR fold, and an electron density map corresponding to an unidentified lipid-like molecule probably derived from the protein expression host is found in the structure. We reveal functions of PTPIP51 in phospholipid binding/transfer, particularly of phosphatidic acid, in vitro. Depletion of PTPIP51 in cells reduces the mitochondrial cardiolipin level. Additionally, we confirm that the PTPIP51-VAPB interaction is mediated by the FFAT-like motif of PTPIP51 and the MSP domain of VAPB. Our findings suggest that PTPIP51 is a phospholipid transfer protein with a MAM-tethering function.


Subject(s)
Calcium , Phospholipids , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phospholipids/metabolism , Protein Tyrosine Phosphatases
8.
Molecules ; 26(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802888

ABSTRACT

Epigenetic regulation is known to play a key role in progression of anti-cancer therapeutics. Lysine acetylation is an important mechanism in controlling gene expression. There has been increasing interest in bromodomain owing to its ability to modulate transcription of various genes as an epigenetic 'reader.' Herein, we report the design, synthesis, and X-ray studies of novel aristoyagonine (benzo[6,7]oxepino[4,3,2-cd]isoindol-2(1H)-one) derivatives and investigate their inhibitory effect against Brd4 bromodomain. Five compounds 8ab, 8bc, 8bd, 8be, and 8bf have been discovered with high binding affinity over the Brd4 protein. Co-crystal structures of these five inhibitors with human Brd4 bromodomain demonstrated that it has a key binding mode occupying the hydrophobic pocket, which is known to be the acetylated lysine binding site. These novel Brd4 bromodomain inhibitors demonstrated impressive inhibitory activity and mode of action for the treatment of cancer diseases.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Enzyme Inhibitors/chemistry , Isoquinolines/chemistry , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Acetylation , Binding Sites/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Isoquinolines/chemical synthesis , Lysine/chemistry , Lysine/metabolism , Protein Binding , Protein Domains/genetics , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Int J Mol Sci ; 21(21)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33114206

ABSTRACT

Aberrant tyrosine-protein kinase Mer (MerTK) expression triggers prosurvival signaling and contributes to cell survival, invasive motility, and chemoresistance in many kinds of cancers. In addition, recent reports suggested that MerTK could be a primary target for abnormal platelet aggregation. Consequently, MerTK inhibitors may promote cancer cell death, sensitize cells to chemotherapy, and act as new antiplatelet agents. We screened an inhouse chemical library to discover novel small-molecule MerTK inhibitors, and identified AZD7762, which is known as a checkpoint-kinase (Chk) inhibitor. The inhibition of MerTK by AZD7762 was validated using an in vitro homogeneous time-resolved fluorescence (HTRF) assay and through monitoring the decrease in phosphorylated MerTK in two lung cancer cell lines. We also determined the crystal structure of the MerTK:AZD7762 complex and revealed the binding mode of AZD7762 to MerTK. Structural information from the MerTK:AZD7762 complex and its comparison with other MerTK:inhibitor structures gave us new insights for optimizing the development of inhibitors targeting MerTK.


Subject(s)
Lung Neoplasms/metabolism , Thiophenes/chemistry , Thiophenes/pharmacology , Urea/analogs & derivatives , c-Mer Tyrosine Kinase/chemistry , c-Mer Tyrosine Kinase/metabolism , A549 Cells , Cell Line, Tumor , Crystallography, X-Ray , Down-Regulation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Models, Molecular , Phosphorylation/drug effects , Protein Binding , Protein Conformation , Protein Domains , Structure-Activity Relationship , Urea/chemistry , Urea/pharmacology
10.
Biochem Biophys Res Commun ; 526(1): 35-40, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32192768

ABSTRACT

Endonuclease G (EndoG) is a mitochondrial enzyme that responds to apoptotic stimuli by translocating to the nucleus and cleaving the chromatin DNA. The molecular mechanism of EndoG still remains unknown in higher organisms. Here, we determined the crystal structure of mouse EndoG at ∼1.96 Å resolution. The EndoG shows an altered dimeric configuration in which N-terminal region of one subunit interact to the other subunit in dimer. The deletion of this region that is highly conserved in mammalian EndoGs resulted in a monomer with significantly reduced activity suggesting the association of the dimeric arrangement into the nuclease activity. Furthermore, we observed a large conformational change in the loop of the active site groove in EndoG, which corresponds to the DNA binding region. Intriguingly, EndoG dimers are linked by oxidation of the reactive cysteine 110 in this flexible loop to form a long oligomeric chain in the crystal lattice. The structural analysis and ensuing biochemical data suggest that this flexible loop region in the active site is important to the regulation of EndoG nuclease function in mouse.


Subject(s)
Endodeoxyribonucleases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Cysteine/metabolism , DNA/chemistry , Mice , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Multimerization
11.
Nat Commun ; 9(1): 1200, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572471

ABSTRACT

BAFF, a member of the TNF superfamily, has been recognized as a good target for autoimmune diseases. Belimumab, an anti-BAFF monoclonal antibody, was approved by the FDA for use in treating systemic lupus erythematosus. However, the molecular basis of BAFF neutralization by belimumab remains unclear. Here our crystal structure of the BAFF-belimumab Fab complex shows the precise epitope and the BAFF-neutralizing mechanism of belimumab, and demonstrates that the therapeutic activity of belimumab involves not only antagonizing the BAFF-receptor interaction, but also disrupting the formation of the more active BAFF 60-mer to favor the induction of the less active BAFF trimer through interaction with the flap region of BAFF. In addition, the belimumab HCDR3 loop mimics the DxL(V/L) motif of BAFF receptors, thereby binding to BAFF in a similar manner as endogenous BAFF receptors. Our data thus provides insights for the design of new drugs targeting BAFF for the treatment of autoimmune diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Cell Activating Factor/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Amino Acid Motifs , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/antagonists & inhibitors , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/immunology , Chromatography, Gel , Crystallography, X-Ray , Epitopes/chemistry , Humans , Immunosuppressive Agents/pharmacology , Ligands , Mutation , Protein Binding
12.
Pharmacol Res ; 128: 211-219, 2018 02.
Article in English | MEDLINE | ID: mdl-29106959

ABSTRACT

Dual specificity phosphatases (DUSPs) include MAP kinase phosphatases and atypical dual specificity phosphatases and mediate cell growth and differentiation, brain function, and immune responses. They serve as targets for drug development against cancers, diabetes and depression. Several DUSPs have non-canonical conformation of the central ß-sheet and active site loops, suggesting that they may have conformational switch that is related to the regulation of enzyme activity. Here, we determined the crystal structure of DUSP13a, and identified two different structures that represent intermediates of the postulated conformational switch. Amino acid sequence of DUSP13a is not significantly homologous to DUSPs with conformational switch, indicating that the conformational switch is not sequence-dependent, but rather determined by ligand interaction. The sequence-independency suggests that other DUSPs with canonical conformation may have the conformational switch during specific cellular regulation. The conformational switch leads to significant changes in the protein surface, including a hydrophobic surface and pockets, which can be exploited for development of allosteric modulators of drug target DUSPs.


Subject(s)
Dual-Specificity Phosphatases , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Protein Conformation
13.
PLoS One ; 12(11): e0187701, 2017.
Article in English | MEDLINE | ID: mdl-29121083

ABSTRACT

Dual-specificity phosphatases (DUSPs) constitute a subfamily of protein tyrosine phosphatases, and are intimately involved in the regulation of diverse parameters of cellular signaling and essential biological processes. DUSP28 is one of the DUSP subfamily members that is known to be implicated in the progression of hepatocellular and pancreatic cancers, and its biological functions and enzymatic characteristics are mostly unknown. Herein, we present the crystal structure of human DUSP28 determined to 2.1 Å resolution. DUSP28 adopts a typical DUSP fold, which is composed of a central ß-sheet covered by α-helices on both sides and contains a well-ordered activation loop, as do other enzymatically active DUSP proteins. The catalytic pocket of DUSP28, however, appears hardly accessible to a substrate because of the presence of nonconserved bulky residues in the protein tyrosine phosphatase signature motif. Accordingly, DUSP28 showed an atypically low phosphatase activity in the biochemical assay, which was remarkably improved by mutations of two nonconserved residues in the activation loop. Overall, this work reports the structural and biochemical basis for understanding a putative oncological therapeutic target, DUSP28, and also provides a unique mechanism for the regulation of enzymatic activity in the DUSP subfamily proteins.


Subject(s)
Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Phosphorylation , Tyrosine
14.
Sci Rep ; 7(1): 5532, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28717238

ABSTRACT

In 2016 and 2017, monoclonal antibodies targeting PD-L1, including atezolizumab, durvalumab, and avelumab, were approved by the FDA for the treatment of multiple advanced cancers. And many other anti-PD-L1 antibodies are under clinical trials. Recently, the crystal structures of PD-L1 in complex with BMS-936559 and avelumab have been determined, revealing details of the antigen-antibody interactions. However, it is still unknown how atezolizumab and durvalumab specifically recognize PD-L1, although this is important for investigating novel binding sites on PD-L1 targeted by other therapeutic antibodies for the design and improvement of anti-PD-L1 agents. Here, we report the crystal structures of PD-L1 in complex with atezolizumab and durvalumab to elucidate the precise epitopes involved and the structural basis for PD-1/PD-L1 blockade by these antibodies. A comprehensive comparison of PD-L1 interactions with anti-PD-L1 antibodies provides a better understanding of the mechanism of PD-L1 blockade as well as new insights into the rational design of improved anti-PD-L1 therapeutics.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/chemistry , Programmed Cell Death 1 Receptor/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Binding Sites , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Humans , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Structure, Quaternary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment
15.
Nat Commun ; 7: 13354, 2016 10 31.
Article in English | MEDLINE | ID: mdl-27796306

ABSTRACT

Cancer cells express tumour-specific antigens derived via genetic and epigenetic alterations, which may be targeted by T-cell-mediated immune responses. However, cancer cells can avoid immune surveillance by suppressing immunity through activation of specific inhibitory signalling pathways, referred to as immune checkpoints. In recent years, the blockade of checkpoint molecules such as PD-1, PD-L1 and CTLA-4, with monoclonal antibodies has enabled the development of breakthrough therapies in oncology, and four therapeutic antibodies targeting these checkpoint molecules have been approved by the FDA for the treatment of several types of cancer. Here, we report the crystal structures of checkpoint molecules in complex with the Fab fragments of therapeutic antibodies, including PD-1/pembrolizumab, PD-1/nivolumab, PD-L1/BMS-936559 and CTLA-4/tremelimumab. These complex structures elucidate the precise epitopes of the antibodies and the molecular mechanisms underlying checkpoint blockade, providing useful information for the improvement of monoclonal antibodies capable of attenuating checkpoint signalling for the treatment of cancer.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal, Humanized/chemistry , Crystallography, X-Ray , Epitopes/chemistry , Humans , Mice , Models, Molecular , Molecular Conformation , Nivolumab , Programmed Cell Death 1 Receptor/chemistry , Protein Binding , Signal Transduction
16.
Nucleic Acids Res ; 44(W1): W416-23, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27151195

ABSTRACT

Many applications, such as protein design, homology modeling, flexible docking, etc. require the prediction of a protein's optimal side-chain conformations from just its amino acid sequence and backbone structure. Side-chain prediction (SCP) is an NP-hard energy minimization problem. Here, we present BetaSCPWeb which efficiently computes a conformation close to optimal using a geometry-prioritization method based on the Voronoi diagram of spherical atoms. Its outputs are visual, textual and PDB file format. The web server is free and open to all users at http://voronoi.hanyang.ac.kr/betascpweb with no login requirement.


Subject(s)
Internet , Mathematics , Proteins/chemistry , Software , Algorithms , Amino Acid Sequence , Databases, Protein , Models, Molecular , Protein Conformation , Thermodynamics
17.
Bioorg Med Chem Lett ; 26(1): 87-93, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26602279

ABSTRACT

Protein tyrosine phosphatase sigma (PTPσ) is a potential target for the therapeutic treatment of neurological deficits associated with impaired neuronal recovery, as this protein is the receptor for chondroitin sulfate proteoglycan (CSPG), which is known to inhibit neuronal regeneration. Through a high-throughput screening approach started from 6400 representative compounds in the Korea Chemical Bank chemical library, we identified 11 novel PTPσ inhibitors that can be classified as flavonoid derivatives or analogs, with IC50 values ranging from 0.5 to 17.5µM. Biochemical assays and structure-based active site-docking simulation indicate that our inhibitors are accommodated at the catalytic active site of PTPσ as surrogates for the phosphotyrosine group. Treatments of these compounds on PC-12 neuronal cells led to the recovery of neurite extension attenuated by CSPG treatment, demonstrating their potential as antineurodegenerative agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Neurites/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 2/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Flavonoids/chemical synthesis , Flavonoids/chemistry , Models, Molecular , Molecular Structure , Neurites/metabolism , PC12 Cells , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
18.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 7): 1528-39, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26143924

ABSTRACT

Myotubularin-related proteins are a large family of phosphoinositide phosphatases; their activity, stability and subcellular localization are regulated by dimeric interactions with other members of the family. Here, the crystal structure of the phosphatase domain of MTMR8 is reported. Conformational deviation of the two loops that mediate interaction with the PH-GRAM domain suggests that the PH-GRAM domain interacts differently with the phosphatase domain of each MTMR member. The protein exists as a dimer with twofold symmetry, providing insight into a novel mode of dimerization mediated by the phosphatase domain. Structural comparison and mutation studies suggest that Lys255 of MTMR8 interacts with the substrate diacylglycerol moiety, similar to Lys333 of MTMR2, although the positions of these residues are different. The catalytic activity of the MTMR8 phosphatase domain is inhibited by oxidation and is reversibly reactivated by reduction, suggesting the presence of an oxidation-protective intermediate other than a disulfide bond owing to the absence of a cysteine within a disulfide-bond distance from Cys338.


Subject(s)
Phosphatidylinositols/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Oxidation-Reduction , Protein Conformation , Protein Multimerization , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Substrate Specificity
19.
Nucleic Acids Res ; 43(W1): W413-8, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25904629

ABSTRACT

Molecular cavities, which include voids and channels, are critical for molecular function. We present a webserver, BetaCavityWeb, which computes these cavities for a given molecular structure and a given spherical probe, and reports their geometrical properties: volume, boundary area, buried area, etc. The server's algorithms are based on the Voronoi diagram of atoms and its derivative construct: the beta-complex. The correctness of the computed result and computational efficiency are both mathematically guaranteed. BetaCavityWeb is freely accessible at the Voronoi Diagram Research Center (VDRC) (http://voronoi.hanyang.ac.kr/betacavityweb).


Subject(s)
Molecular Conformation , Software , Algorithms , Internet , Models, Molecular , Protein Conformation
20.
FEBS Lett ; 589(2): 193-200, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25481411

ABSTRACT

To improve a previously constructed broadly neutralizing hepatitis B virus (HBV)-specific preS1 humanized antibody (HzKR127), we further humanized it through specificity-determining residue (SDR) grafting. Moreover, we improved affinity by mutating two residues in heavy-chain complementarity-determining regions (CDR), on the basis of the crystal structure of the antigen-antibody complex. HzKR127-3.2 exhibited 2.5-fold higher affinity and enhanced virus-neutralizing activity compared to the original KR127 antibody and showed less immunogenic potential than HzKR127. Enhanced virus-neutralizing activity was achieved by the increased association rate, providing insights into engineering potent antibody therapeutics for HBV immunoprophylaxis. HzKR127-3.2 may be a good candidate for HBV immunoprophylaxis.


Subject(s)
Antigen-Antibody Complex , Hepatitis B Antibodies/chemistry , Hepatitis B Antibodies/immunology , Hepatitis B virus/immunology , Amino Acid Sequence , Animals , Cell Line , Epitopes, T-Lymphocyte/immunology , Hepatitis B Antibodies/pharmacology , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Viral Proteins/chemistry , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...