Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2705: 199-210, 2023.
Article in English | MEDLINE | ID: mdl-37668975

ABSTRACT

Biosensor measurement using surface plasmon resonance enables precise evaluation of peptide-protein interactions. It is a sensitive technique that provides kinetic and affinity data with very little sample and without the need for analyte labels. Here, we describe its application for the analysis of peptide interactions with the Grb7-SH2 domain prepared with a GST-tag for tethering to the chip surface. This has been successfully and reliably used for direct comparison of a range of peptides under different solution conditions as well as direct comparison of peptides flowed over different related SH2 domains in real time. We have used the BIAcore system and describe both the methodology for data collection and analysis, with principles also applicable to other biosensor platforms.


Subject(s)
Surface Plasmon Resonance , src Homology Domains , Data Collection , Kinetics , Peptides
2.
J Med Chem ; 66(8): 5859-5872, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37071570

ABSTRACT

The development of low-affinity fragment hits into higher-affinity leads is a major hurdle in fragment-based drug design. Here, we demonstrate the Rapid Elaboration of Fragments into Leads (REFiL) by applying an integrated workflow that provides a systematic approach to generate higher-affinity binders without the need for structural information. The workflow involves the selection of commercial analogues of fragment hits to generate preliminary structure-activity relationships. This is followed by parallel microscale chemistry using chemoinformatically designed reagent libraries to rapidly explore chemical diversity. After a fragment screen against bromodomain-3 extra-terminal (BRD3-ET) domain, we applied the REFiL workflow, which allowed us to develop a series of ligands that bind to BRD3-ET. With REFiL, we were able to rapidly improve binding affinity > 30-fold. REFiL can be applied readily to a broad range of proteins without the need for a structure, allowing the efficient evolution of low-affinity fragments into higher-affinity leads and chemical probes.


Subject(s)
Drug Design , Proteins , Proteins/metabolism , Structure-Activity Relationship , Protein Domains , Ligands
3.
Molecules ; 28(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37049868

ABSTRACT

Human immunodeficiency virus type I (HIV-1) is a retrovirus that infects cells of the host's immune system leading to acquired immunodeficiency syndrome and potentially death. Although treatments are available to prevent its progression, HIV-1 remains a major burden on health resources worldwide. Continued emergence of drug-resistance mutations drives the need for novel drugs that can inhibit HIV-1 replication through new pathways. The viral protein reverse transcriptase (RT) plays a fundamental role in the HIV-1 replication cycle, and multiple approved medications target this enzyme. In this study, fragment-based drug discovery was used to optimize a previously identified hit fragment (compound B-1), which bound RT at a novel site. Three series of compounds were synthesized and evaluated for their HIV-1 RT binding and inhibition. These series were designed to investigate different vectors around the initial hit in an attempt to improve inhibitory activity against RT. Our results show that the 4-position of the core scaffold is important for binding of the fragment to RT, and a lead compound with a cyclopropyl substitution was selected and further investigated. Requirements for binding to the NNRTI-binding pocket (NNIBP) and a novel adjacent site were investigated, with lead compound 27-a minimal but efficient NNRTI-offering a starting site for the development of novel dual NNIBP-Adjacent site inhibitors.


Subject(s)
Acquired Immunodeficiency Syndrome , Anti-HIV Agents , HIV-1 , Humans , Reverse Transcriptase Inhibitors/chemistry , HIV Reverse Transcriptase , Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use
4.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217625

ABSTRACT

As natural chemokine inhibitors, evasin proteins produced in tick saliva are potential therapeutic agents for numerous inflammatory diseases. Engineering evasins to block the desired chemokines and avoid off-target side effects requires structural understanding of their target selectivity. Structures of the class A evasin EVA-P974 bound to human CC chemokine ligands 7 and 17 (CCL7 and CCL17) and to a CCL8-CCL7 chimera reveal that the specificity of class A evasins for chemokines of the CC subfamily is defined by conserved, rigid backbone-backbone interactions, whereas the preference for a subset of CC chemokines is controlled by side-chain interactions at four hotspots in flexible structural elements. Hotspot mutations alter target preference, enabling inhibition of selected chemokines. The structure of an engineered EVA-P974 bound to CCL2 reveals an underlying molecular mechanism of EVA-P974 target preference. These results provide a structure-based framework for engineering evasins as targeted antiinflammatory therapeutics.


Subject(s)
Arthropod Proteins/chemistry , Chemokines/metabolism , Inflammation/metabolism , Protein Engineering , Ticks/metabolism , Animals , Arthropod Proteins/metabolism , Protein Binding , Protein Conformation , Receptors, Chemokine/metabolism
5.
Nucleic Acids Res ; 49(5): 2403-2417, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33621982

ABSTRACT

TIA-1 is an RNA-binding protein that sequesters target RNA into stress granules under conditions of cellular stress. Promotion of stress granule formation by TIA-1 depends upon self-association of its prion-like domain that facilitates liquid-liquid phase separation and is thought to be enhanced via RNA binding. However, the mechanisms underlying the influence of RNA on TIA-1 self-association have not been previously demonstrated. Here we have investigated the self-associating properties of full-length TIA-1 in the presence of designed and native TIA-1 nucleic acid binding sites in vitro, monitoring phase separation, fibril formation and shape. We show that single stranded RNA and DNA induce liquid-liquid phase separation of TIA-1 in a multisite, sequence-specific manner and also efficiently promote formation of amyloid-like fibrils. Although RNA binding to a single site induces a small conformational change in TIA-1, this alone does not enhance phase separation of TIA-1. Tandem binding sites are required to enhance phase separation of TIA-1 and this is finely tuned by the protein:binding site stoichiometry rather than nucleic acid length. Native tandem TIA-1 binding sites within the 3' UTR of p53 mRNA also efficiently enhance phase separation of TIA-1 and thus may potentially act as potent nucleation sites for stress granule assembly.


Subject(s)
RNA/metabolism , T-Cell Intracellular Antigen-1/chemistry , 3' Untranslated Regions , Amyloid/ultrastructure , Binding Sites , DNA/chemistry , DNA/metabolism , Humans , Models, Molecular , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Protein Conformation , RNA/chemistry , T-Cell Intracellular Antigen-1/metabolism , T-Cell Intracellular Antigen-1/ultrastructure , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Cell Rep ; 28(8): 2023-2036.e4, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433980

ABSTRACT

Lymphatic vascular development establishes embryonic and adult tissue fluid balance and is integral in disease. In diverse vertebrate organs, lymphatic vessels display organotypic function and develop in an organ-specific manner. In all settings, developmental lymphangiogenesis is considered driven by vascular endothelial growth factor (VEGF) receptor-3 (VEGFR3), whereas a role for VEGFR2 remains to be fully explored. Here, we define the zebrafish Vegf/Vegfr code in receptor binding studies. We find that while Vegfd directs craniofacial lymphangiogenesis, it binds Kdr (a VEGFR2 homolog) but surprisingly, unlike in mammals, does not bind Flt4 (VEGFR3). Epistatic analyses and characterization of a kdr mutant confirm receptor-binding analyses, demonstrating that Kdr is indispensible for rostral craniofacial lymphangiogenesis, but not caudal trunk lymphangiogenesis, in which Flt4 is central. We further demonstrate an unexpected yet essential role for Kdr in inducing lymphatic endothelial cell fate. This work reveals evolutionary divergence in the Vegf/Vegfr code that uncovers spatially restricted mechanisms of developmental lymphangiogenesis.


Subject(s)
Endothelial Cells/metabolism , Evolution, Molecular , Lymphangiogenesis , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Ligands , Mice , Protein Binding , Proteolysis , Reproducibility of Results , Vascular Endothelial Growth Factor C/chemistry , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
7.
Elife ; 72018 12 21.
Article in English | MEDLINE | ID: mdl-30575518

ABSTRACT

Proper regulation of germline gene expression is essential for fertility and maintaining species integrity. In the C. elegans germline, a diverse repertoire of regulatory pathways promote the expression of endogenous germline genes and limit the expression of deleterious transcripts to maintain genome homeostasis. Here we show that the conserved TRIM-NHL protein, NHL-2, plays an essential role in the C. elegans germline, modulating germline chromatin and meiotic chromosome organization. We uncover a role for NHL-2 as a co-factor in both positively (CSR-1) and negatively (HRDE-1) acting germline 22G-small RNA pathways and the somatic nuclear RNAi pathway. Furthermore, we demonstrate that NHL-2 is a bona fide RNA binding protein and, along with RNA-seq data point to a small RNA independent role for NHL-2 in regulating transcripts at the level of RNA stability. Collectively, our data implicate NHL-2 as an essential hub of gene regulatory activity in both the germline and soma.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Carrier Proteins/metabolism , Germ Cells/metabolism , RNA Interference , Animals , Chromatin/metabolism , Gene Regulatory Networks
8.
Front Mol Biosci ; 4: 64, 2017.
Article in English | MEDLINE | ID: mdl-29018805

ABSTRACT

Growth factor receptor bound protein 7 (Grb7) is an adaptor protein with established roles in the progression of both breast and pancreatic cancers. Through its C-terminal SH2 domain, Grb7 binds to phosphorylated tyrosine kinases to promote proliferative and migratory signaling. Here, we investigated the molecular basis for the specificity of a Grb7 SH2-domain targeted peptide inhibitor. We identified that arginine 462 in the BC loop is unique to Grb7 compared to Grb2, another SH2 domain bearing protein that shares the same consensus binding motif as Grb7. Using surface plasmon resonance we demonstrated that Grb7-SH2 binding to G7-18NATE is reduced 3.3-fold when the arginine is mutated to the corresponding Grb2 amino acid. The reverse mutation in Grb2-SH2 (serine to arginine), however, was insufficient to restore binding of G7-18NATE to Grb2-SH2. Further, using a microarray, we confirmed that G7-18NATE is specific for Grb7 over a panel of 79 SH2 domains, and identified that leucine at the ßD6 position may also be a requirement for Grb7-SH2 binding. This study provides insight into the specificity defining features of Grb7 for the inhibitor molecule G7-18NATE, that will assist in the development of improved Grb7 targeted inhibitors.

9.
J Med Chem ; 60(22): 9349-9359, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29083893

ABSTRACT

Grb7 is a signaling protein with critical roles in tumor cell proliferation and migration and an established cancer therapeutic target. Here we explore chemical space to develop a new bicyclic peptide inhibitor, incorporating thioether and lactam linkers that binds with affinity (KD = 1.1 µM) and specificity to the Grb7-SH2 domain. Structural analysis of the Grb7-SH2/peptide complex revealed an unexpected binding orientation underlying the binding selectivity by this new scaffold. We further incorporated carboxymethylphenylalanine and carboxyphenylalanine phosphotyrosine mimetics and arrived at an optimized inhibitor that potently binds Grb7-SH2 (KD = 0.13 µM) under physiological conditions. X-ray crystal structures of these Grb7-SH2/peptide complexes reveal the structural basis for the most potent and specific inhibitors of Grb7 developed to date. Finally, we demonstrate that cell permeable versions of these peptides successfully block Grb7 mediated interactions in a breast cancer cell line, establishing the potential of these peptides in the development of novel therapeutics targeted to Grb7.


Subject(s)
GRB7 Adaptor Protein/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Focal Adhesion Kinase 1/metabolism , GRB7 Adaptor Protein/chemistry , GRB7 Adaptor Protein/metabolism , Humans , Lactams/chemical synthesis , Lactams/chemistry , Lactams/pharmacology , Ligands , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Phosphates/chemistry , Protein Conformation , Receptor, ErbB-2/metabolism , Shc Signaling Adaptor Proteins/metabolism , src Homology Domains
10.
ACS Chem Biol ; 11(8): 2357-65, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27362503

ABSTRACT

Phenol and its derivatives are common pollutants that are present in industrial discharge and are major xenobiotics that lead to water pollution. To monitor as well as improve water quality, attempts have been made in the past to engineer bacterial in vivo biosensors. However, due to the paucity of structural information, there is insufficiency in gauging the factors that lead to high sensitivity and selectivity, thereby impeding development. Here, we present the crystal structure of the sensor domain of MopR (MopR(AB)) from Acinetobacter calcoaceticus in complex with phenol and its derivatives to a maximum resolution of 2.5 Å. The structure reveals that the N-terminal residues 21-47 possess a unique fold, which are involved in stabilization of the biological dimer, and the central ligand binding domain belongs to the "nitric oxide signaling and golgi transport" fold, commonly present in eukaryotic proteins that bind long-chain fatty acids. In addition, MopR(AB) nests a zinc atom within a novel zinc binding motif, crucial for maintaining structural integrity. We propose that this motif is crucial for orchestrated motions associated with the formation of the effector binding pocket. Our studies reveal that residues W134 and H106 play an important role in ligand binding and are the key selectivity determinants. Furthermore, comparative analysis of MopR with XylR and DmpR sensor domains enabled the design of a MopR binding pocket that is competent in binding DmpR-specific ligands. Collectively, these findings pave way towards development of specific/broad based biosensors, which can act as useful tools for detection of this class of pollutants.


Subject(s)
Bacterial Proteins/metabolism , Environmental Pollutants/analysis , Gene Expression Regulation, Bacterial , Trans-Activators/metabolism , Acinetobacter/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biosensing Techniques , Crystallography, X-Ray , Dimerization , Ligands , Trans-Activators/chemistry , Trans-Activators/genetics
11.
Sci Rep ; 6: 27060, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27257138

ABSTRACT

The design of potent and specific peptide inhibitors to therapeutic targets is of enormous utility for both proof-of-concept studies and for the development of potential new therapeutics. Grb7 is a key signaling molecule in the progression of HER2 positive and triple negative breast cancers. Here we report the crystal structure of a stapled bicyclic peptide inhibitor G7-B1 in complex with the Grb7-SH2 domain. This revealed an unexpected binding mode of the peptide, in which the staple forms an alternative contact with the surface of the target protein. Based on this structural information, we designed a new series of bicyclic G7 peptides that progressively constrain the starting peptide, to arrive at the G7-B4 peptide that binds with an approximately 2-fold enhanced affinity to the Grb7-SH2 domain (KD = 0.83 µM) compared to G7-B1 and shows low affinity binding to Grb2-, Grb10- and Grb14-SH2 domains (KD > 100 µM). Furthermore, we determined the structure of the G7-B4 bicyclic peptide in complex with the Grb7-SH2 domain, both before and after ring closing metathesis to show that the closed staple is essential to the target interaction. The G7-B4 peptide represents an advance in the development of Grb7 inhibitors and is a classical example of structure aided inhibitor development.


Subject(s)
GRB7 Adaptor Protein/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Drug Design , GRB7 Adaptor Protein/antagonists & inhibitors , Humans , Hydrogen Bonding , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary
12.
J Struct Biol ; 194(2): 205-13, 2016 May.
Article in English | MEDLINE | ID: mdl-26923153

ABSTRACT

Chemotaxis and motility play an important role in the colonisation of avian and human hosts by Campylobacter jejuni. Chemotactic recognition of extracellular signals is mediated by the periplasmic sensing domain of methyl-accepting chemotactic proteins (membrane-embedded receptors). In this work, we report a high-resolution structure of the periplasmic sensing domain of transducer-like protein 1 (Tlp1), an aspartate receptor of C. jejuni. Crystallographic analysis revealed that it contains two Per-Arnt-Sim (PAS) subdomains. An acetate and chloride ions (both from the crystallisation buffer) were observed bound to the membrane-proximal and membrane-distal PAS subdomains, respectively. Surprisingly, despite being crystallised in the presence of aspartate, the structure did not show any electron density corresponding to this amino acid. Furthermore, no binding between the sensing domain of Tlp1 and aspartate was detected by microcalorimetric experiments. These structural and biophysical data suggest that Tlp1 does not sense aspartate directly; instead, ligand recognition is likely to occur indirectly via an as yet unidentified periplasmic binding protein.


Subject(s)
Aspartic Acid/chemistry , Bacterial Proteins/chemistry , Campylobacter jejuni/chemistry , Receptors, Amino Acid/chemistry , Aspartic Acid/metabolism , Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Chemotaxis/physiology , Crystallography, X-Ray , Ligands , Models, Molecular , Protein Domains , Protein Structure, Secondary , Receptors, Amino Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
13.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 10): 2127-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26457436

ABSTRACT

Chemotaxis, mediated by methyl-accepting chemotaxis protein (MCP) receptors, plays an important role in the ecology of bacterial populations. This paper presents the first crystallographic analysis of the structure and ligand-induced conformational changes of the periplasmic tandem Per-Arnt-Sim (PAS) sensing domain (PTPSD) of a characterized MCP chemoreceptor. Analysis of the complex of the Campylobacter jejuni Tlp3 PTPSD with isoleucine (a chemoattractant) revealed that the PTPSD is a dimer in the crystal. The two ligand-binding sites are located in the membrane-distal PAS domains on the faces opposite to the dimer interface. Mutagenesis experiments show that the five strongly conserved residues that stabilize the main-chain moiety of isoleucine are essential for binding, suggesting that the mechanism by which this family of chemoreceptors recognizes amino acids is highly conserved. Although the fold and mode of ligand binding of the PTPSD are different from the aspartic acid receptor Tar, the structural analysis suggests that the PTPSDs of amino-acid chemoreceptors are also likely to signal by a piston displacement mechanism. The PTPSD fluctuates between piston (C-terminal helix) `up' and piston `down' states. Binding of an attractant to the distal PAS domain locks it in the closed form, weakening its association with the proximal domain and resulting in the transition of the latter into an open form, concomitant with a downward (towards the membrane) 4 Špiston displacement of the C-terminal helix. In vivo, this movement would generate a transmembrane signal by driving a downward displacement of the transmembrane helix 2 towards the cytoplasm.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Isoleucine/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Binding Sites , Campylobacter Infections/microbiology , Campylobacter jejuni/chemistry , Campylobacter jejuni/cytology , Chemotaxis , Crystallography, X-Ray , Isoleucine/chemistry , Membrane Proteins/chemistry , Methyl-Accepting Chemotaxis Proteins , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction
14.
J Med Chem ; 58(19): 7707-18, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26359549

ABSTRACT

The Grb7 adaptor protein is a therapeutic target for both TNBC and HER2+ breast cancer. A nonphosphorylated cyclic peptide 1 (known as G7-18NATE) inhibits Grb7 via targeting the Grb7-SH2 domain, but requires the presence of phosphate ions for both affinity and specificity. Here we report the discovery of malonate bound in the phosphotyrosine binding pocket of the apo-Grb7-SH2 structure. Based on this, we carried out the rational design and synthesis of two analogues of peptide 1 that incorporate carboxymethylphenylalanine (cmF) and carboxyphenylalanine (cF) as mimics of phosphotyrosine (pY). Binding studies using SPR confirmed that affinity for Grb7-SH2 domain is improved up to 9-fold over peptide 1 under physiological phosphate conditions (KD = 2.1-5.7 µM) and that binding is specific for Grb7-SH2 over closely related domains (low or no detectable binding to Grb2-SH2 and Grb10-SH2). X-ray crystallographic structural analysis of the analogue bearing a cmF moiety in complex with Grb7-SH2 has identified the precise contacts conferred by the pY mimic that underpin this improved molecular interaction. Together this study identifies and characterizes the tightest specific inhibitor of Grb7 to date, representing a significant development toward a new Grb7-targeted therapeutic.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , GRB7 Adaptor Protein/antagonists & inhibitors , Peptides, Cyclic/chemistry , Phosphotyrosine/chemistry , Antineoplastic Agents/chemical synthesis , Binding Sites , Breast Neoplasms/drug therapy , Crystallography, X-Ray , Female , GRB7 Adaptor Protein/metabolism , Humans , Malonates/chemistry , Molecular Targeted Therapy , Peptides, Cyclic/chemical synthesis , Peptidomimetics , Phosphates/chemistry , Phosphates/metabolism , Protein Conformation , src Homology Domains
15.
Mol Immunol ; 67(2 Pt B): 287-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26130224

ABSTRACT

The interaction between mannose-binding lectin [MBL]-associated serine protease-2 (MASP-2) and its first substrate, C4 is crucial to the lectin pathway of complement, which is vital for innate host immunity, but also involved in a number of inflammatory diseases. Recent data suggests that two areas outside of the active site of MASP-2 (so-called exosites) are crucial for efficient cleavage of C4: one at the junction of the two complement control protein (CCP) domains of the enzyme and the second on the serine protease (SP) domain. Here, we have further investigated the roles of each of these exosites in the binding and cleavage of C4. We have found that both exosites are required for high affinity binding and efficient cleavage of the substrate protein. Within the SP domain exosite, we have shown here that two arginine residues are most important for high affinity binding and efficient cleavage of C4. Finally, we show that the CCP domain exosite appears to play the major role in the initial interaction with C4, whilst the SP domain exosite plays the major role in a secondary conformational change between the two proteins required to form a high affinity complex. This data has provided new insights into the binding and cleavage of C4 by MASP-2, which may be useful in the design of molecules that modulate this important interaction required to activate the lectin pathway of complement.


Subject(s)
Complement C4/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Immobilized Proteins/metabolism , Kinetics , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance , Time Factors
16.
Cell Cycle ; 14(17): 2729-33, 2015.
Article in English | MEDLINE | ID: mdl-26177105

ABSTRACT

Both RNA-binding proteins (RBP) and miRNA play important roles in the regulation of mRNA expression, often acting together to regulate a target mRNA. In some cases the RBP and miRNA have been reported to act competitively, but in other instances they function cooperatively. Here, we investigated HuR function as an enhancer of let-7-mediated translational repression of c-Myc despite the separation of their binding sites. Using an in vitro system, we determined that a let-7 mimic, consisting of single-stranded (ss)DNA complementary to the let-7 binding site, enhanced the affinity of HuR for a 122-nt MYC RNA encompassing both binding sites. This finding supports the biophysical principle of cooperative binding by an RBP and miRNA purely through interactions at distal mRNA binding sites.


Subject(s)
ELAV-Like Protein 1/metabolism , MicroRNAs/metabolism , Molecular Mimicry/physiology , Proto-Oncogene Proteins c-myc/metabolism , ELAV-Like Protein 1/chemistry , Humans , MicroRNAs/chemistry , Protein Binding/physiology , Proto-Oncogene Proteins c-myc/chemistry
17.
RNA Biol ; 11(6): 766-76, 2014.
Article in English | MEDLINE | ID: mdl-24824036

ABSTRACT

T-cell intracellular antigen-1 (TIA-1) is a key DNA/RNA binding protein that regulates translation by sequestering target mRNAs in stress granules (SG) in response to stress conditions. TIA-1 possesses three RNA recognition motifs (RRM) along with a glutamine-rich domain, with the central domains (RRM2 and RRM3) acting as RNA binding platforms. While the RRM2 domain, which displays high affinity for U-rich RNA sequences, is primarily responsible for interaction with RNA, the contribution of RRM3 to bind RNA as well as the target RNA sequences that it binds preferentially are still unknown. Here we combined nuclear magnetic resonance (NMR) and surface plasmon resonance (SPR) techniques to elucidate the sequence specificity of TIA-1 RRM3. With a novel approach using saturation transfer difference NMR (STD-NMR) to quantify protein-nucleic acids interactions, we demonstrate that isolated RRM3 binds to both C- and U-rich stretches with micromolar affinity. In combination with RRM2 and in the context of full-length TIA-1, RRM3 significantly enhanced the binding to RNA, particularly to cytosine-rich RNA oligos, as assessed by biotinylated RNA pull-down analysis. Our findings provide new insight into the role of RRM3 in regulating TIA-1 binding to C-rich stretches, that are abundant at the 5' TOPs (5' terminal oligopyrimidine tracts) of mRNAs whose translation is repressed under stress situations.


Subject(s)
Nucleotide Motifs , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/metabolism , Protein Interaction Domains and Motifs , RNA/chemistry , RNA/genetics , Base Sequence , Binding Sites , GC Rich Sequence , Humans , Nuclear Magnetic Resonance, Biomolecular , Position-Specific Scoring Matrices
18.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 2): 182-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24637751

ABSTRACT

Human growth factor receptor-bound protein 7 (Grb7) is an adapter protein involved in cell growth, migration and proliferation. It is now recognized that Grb7 is an emerging therapeutic target in specific cancer subtypes. Recently, the discovery of a bicyclic peptide inhibitor that targets the Grb7 SH2 domain, named G7-B1, was reported. In an attempt to probe the foundation of its interaction with Grb7, the crystallization and preliminary data collection of both the apo and G7-B1-bound forms of the Grb7 SH2 domain are reported here. Diffraction-quality crystals were obtained using the hanging-drop vapour-diffusion method. After several rounds of microseeding, crystals of the apo Grb7 SH2 domain were obtained that diffracted to 1.8 Šresolution, while those of the G7-B1-Grb7 SH2 domain complex diffracted to 2.2 Šresolution. The apo Grb7 SH2 domain crystallized in the trigonal space group P63, whereas the G7-B1-Grb7 SH2 domain complex crystallized in the monoclinic space group P21. The experimental aspects of crystallization, crystal optimization and data collection and the preliminary data are reported.


Subject(s)
GRB7 Adaptor Protein/chemistry , Peptides, Cyclic/chemistry , src Homology Domains , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Humans , Protein Conformation
19.
J Biol Chem ; 288(22): 15821-9, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23592783

ABSTRACT

The complement system is an ancient innate immune defense pathway that plays a front line role in eliminating microbial pathogens. Recognition of foreign targets by antibodies drives sequential activation of two serine proteases, C1r and C1s, which reside within the complement Component 1 (C1) complex. Active C1s propagates the immune response through its ability to bind and cleave the effector molecule complement Component 4 (C4). Currently, the precise structural and biochemical basis for the control of the interaction between C1s and C4 is unclear. Here, using surface plasmon resonance, we show that the transition of the C1s zymogen to the active form is essential for C1s binding to C4. To understand this, we determined the crystal structure of a zymogen C1s construct (comprising two complement control protein (CCP) domains and the serine protease (SP) domain). These data reveal that two loops (492-499 and 573-580) in the zymogen serine protease domain adopt a conformation that would be predicted to sterically abrogate C4 binding. The transition from zymogen to active C1s repositions both loops such that they would be able to interact with sulfotyrosine residues on C4. The structure also shows the junction of the CCP1 and CCP2 domains of C1s for the first time, yielding valuable information about the exosite for C4 binding located at this position. Together, these data provide a structural explanation for the control of the interaction with C1s and C4 and, furthermore, point to alternative strategies for developing therapeutic approaches for controlling activation of the complement cascade.


Subject(s)
Complement C1s/chemistry , Complement C4/chemistry , Enzyme Precursors/chemistry , Complement C1s/genetics , Complement C1s/metabolism , Complement C4/genetics , Complement C4/metabolism , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Humans , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
20.
Biopolymers ; 100(5): 543-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23505041

ABSTRACT

Grb7 is an adapter protein involved in the propagation of signals in cancer cell migration and proliferation, and is thus a target for the development of novel anti-cancer agents. An 11-residue thioether-cyclized peptide known as G7-18NATE has previously been developed, that inhibits Grb7 via specific interactions with its SH2 domain with micromolar affinity. Here we explore whether the peptide binding is enhanced by the addition of a second linkage designed to restrain the peptide in its bound conformation and thus reduce the entropic loss upon binding. The use of an O-ally ser covalent linkage between residue positions 1 and 8 successfully enhanced the affinity, and ITC showed that the entropic loss was reduced. A peptide with thioether-cyclization exchanged for an amide linkage showed reduce affinity, though the formation of a disulfide bond between positions 1 and 8 in this peptide enhanced its binding. This study paves the way for improving the G7-18NATE scaffold for second generation inhibitors of Grb7.


Subject(s)
GRB7 Adaptor Protein , src Homology Domains , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , GRB7 Adaptor Protein/metabolism , Humans , Peptides/pharmacology , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...