Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Biol Chem ; 288(10): 7387-98, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23316052

ABSTRACT

Molecular interactions between the tumor suppressor p53 and the anti-apoptotic Bcl-2 family proteins play an important role in the transcription-independent apoptosis of p53. The p53 transactivation domain (p53TAD) contains two conserved ΦXXΦΦ motifs (Φ indicates a bulky hydrophobic residue and X is any other residue) referred to as p53TAD1 (residues 15-29) and p53TAD2 (residues 39-57). We previously showed that p53TAD1 can act as a binding motif for anti-apoptotic Bcl-2 family proteins. In this study, we have identified p53TAD2 as a binding motif for anti-apoptotic Bcl-2 family proteins by using NMR spectroscopy, and we calculated the structures of Bcl-X(L)/Bcl-2 in complex with the p53TAD2 peptide. NMR chemical shift perturbation data showed that p53TAD2 peptide binds to diverse members of the anti-apoptotic Bcl-2 family independently of p53TAD1, and the binding between p53TAD2 and p53TAD1 to Bcl-X(L) is competitive. Refined structural models of the Bcl-X(L)·p53TAD2 and Bcl-2·p53TAD2 complexes showed that the binding sites occupied by p53TAD2 in Bcl-X(L) and Bcl-2 overlap well with those occupied by pro-apoptotic BH3 peptides. Taken together with the mutagenesis, isothermal titration calorimetry, and paramagnetic relaxation enhancement data, our structural comparisons provided the structural basis of p53TAD2-mediated interaction with the anti-apoptotic proteins, revealing that Bcl-X(L)/Bcl-2, MDM2, and cAMP-response element-binding protein-binding protein/p300 share highly similar modes of binding to the dual p53TAD motifs, p53TAD1 and p53TAD2. In conclusion, our results suggest that the dual-site interaction of p53TAD is a highly conserved mechanism underlying target protein binding in the transcription-dependent and transcription-independent apoptotic pathways of p53.


Subject(s)
Apoptosis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Tumor Suppressor Protein p53/chemistry , Amino Acid Motifs/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Binding Sites/genetics , Binding, Competitive , Calorimetry , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/chemistry , bcl-X Protein/genetics , bcl-X Protein/metabolism
2.
Biomol NMR Assign ; 18(1): 71-78, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38551798

ABSTRACT

The intraflagellar transport (IFT) machinery plays a crucial role in the bidirectional trafficking of components necessary for ciliary signaling, such as the Hedgehog, Wnt/PCR, and cAMP/PKA systems. Defects in some components of the IFT machinery cause dysfunction, leading to a wide range of human diseases and developmental disorders termed ciliopathies, such as nephronophthisis. The IFT machinery comprises three sub-complexes: BBsome, IFT-A, and IFT-B. The IFT protein 54 (IFT54) is an important component of the IFT-B sub-complex. In anterograde movement, IFT54 binds to active kinesin-II, walking along the cilia microtubule axoneme and carrying the dynein-2 complex in an inactive state, which works for retrograde movement. Several mutations in IFT54 are known to cause Senior-Loken syndrome, a ciliopathy. IFT54 possesses a divergent Calponin Homology (CH) domain termed as NN-CH domain at its N-terminus. However, several aspects of the function of the NN-CH domain of IFT54 are still obscure. Here, we report the 1H, 15N, and 13C resonance assignments of the NN-CH domain of human IFT54 and its solution structure. The NN-CH domain of human IFT54 adopts essentially the α1-α2-α3-α4-α5 topology as that of mouse IFT54, whose structure was determined by X-ray crystallographic study. The structural information and assignments obtained in this study shed light on the molecular function of the NN-CH domain in IFT54.


Subject(s)
Microfilament Proteins , Protein Domains , Humans , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calponins , Microfilament Proteins/chemistry , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Solutions
3.
Nucleic Acids Res ; 39(4): 1538-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20926394

ABSTRACT

Human Transformer2-ß (hTra2-ß) is an important member of the serine/arginine-rich protein family, and contains one RNA recognition motif (RRM). It controls the alternative splicing of several pre-mRNAs, including those of the calcitonin/calcitonin gene-related peptide (CGRP), the survival motor neuron 1 (SMN1) protein and the tau protein. Accordingly, the RRM of hTra2-ß specifically binds to two types of RNA sequences [the CAA and (GAA)(2) sequences]. We determined the solution structure of the hTra2-ß RRM (spanning residues Asn110-Thr201), which not only has a canonical RRM fold, but also an unusual alignment of the aromatic amino acids on the ß-sheet surface. We then solved the complex structure of the hTra2-ß RRM with the (GAA)(2) sequence, and found that the AGAA tetra-nucleotide was specifically recognized through hydrogen-bond formation with several amino acids on the N- and C-terminal extensions, as well as stacking interactions mediated by the unusually aligned aromatic rings on the ß-sheet surface. Further NMR experiments revealed that the hTra2-ß RRM recognizes the CAA sequence when it is integrated in the stem-loop structure. This study indicates that the hTra2-ß RRM recognizes two types of RNA sequences in different RNA binding modes.


Subject(s)
Nerve Tissue Proteins/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Guanine/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Serine-Arginine Splicing Factors
4.
Biochem Biophys Res Commun ; 420(1): 48-53, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22402281

ABSTRACT

Multi-targeting therapy is an emerging strategy of drug discovery to improve therapeutic efficacy, safety and resistance profiles. In this study, we monitored the binding of a potent MDM2 inhibitor Nutlin-3 with anti-apoptotic Bcl-2 family proteins using NMR spectroscopy. Our results showed the universal binding of Nutlin-3 with diverse anti-apoptotic Bcl-2 family proteins. Taken together with the binding data for Nutlin-3 analogs, the structural model of the Bcl-X(L)/Nutlin-3 complex showed that the binding mode of Nutlin-3 resembles that of the Bcl-X(L)/Bcl-2 inhibitors, suggesting the molecular mechanism of transcription-independent mitochondrial apoptosis by Nutlin-3. Finally, our structural comparison provides structural insights into the dual-targeting mechanism of how Nutlin-3 can bind to two different target proteins, MDM2 and anti-apoptotic Bcl-2 family proteins in a similar manner.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Apoptosis , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Structure-Activity Relationship , bcl-X Protein/chemistry
5.
Biomol NMR Assign ; 16(1): 41-49, 2022 04.
Article in English | MEDLINE | ID: mdl-34783967

ABSTRACT

Matrin-3 is a multifunctional protein that binds to both DNA and RNA. Its DNA-binding activity is linked to the formation of the nuclear matrix and transcriptional regulation, while its RNA-binding activity is linked to mRNA metabolism including splicing, transport, stabilization, and degradation. Correspondingly, Matrin-3 has two zinc finger domains for DNA binding and two consecutive RNA recognition motif (RRM) domains for RNA binding. Matrin-3 has been reported to cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) when its disordered region contains pathogenic mutations. Simultaneously, it has been shown that the RNA-binding activity of Matrin-3 mediated by its RRM domains, affects the formation of insoluble cytoplasmic granules, which are related to the pathogenic mechanism of ALS/FTD. Thus, the effect of the RRM domains on the phase separation of condensed protein/RNA mixtures has to be clarified for a comprehensive understanding of ALS/FTD. Here, we report the 1H, 15N, and 13C resonance assignments of the two RNA binding domains and their solution structures. The resonance assignments and the solution structures obtained in this work will contribute to the elucidation of the molecular basis of Matrin-3 in the pathogenic mechanism of ALS and/or FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Humans , Nuclear Magnetic Resonance, Biomolecular , RNA/metabolism , RNA Recognition Motif
6.
Biomol NMR Assign ; 15(1): 1-7, 2021 04.
Article in English | MEDLINE | ID: mdl-32930954

ABSTRACT

In humans, YTH (YT521-B homology) domain containing protein 2 (YTHDC2) plays a crucial role in the phase-shift from mitosis to meiosis. YTH domains bind to methylated adenosine nucleotides such as m6A. In a phylogenic tree, the YTH domain of YTHDC2 (YTH2) and that of the YTH containing protein YTHDC1 (YTH1) belong to the same sub-group. However, the binding affinity of m6A differs between these proteins. Here, we report 1H, 13C and 15N resonance assignment of YTH2 and its solution structure to examine the difference of the structural architecture and the dynamic properties of YTH1 and YTH2. YTH2 adopts a ß1-α1-ß2-α2-ß3-ß4-ß5-α3-ß6-α4 topology, which was also observed in YTH1. However, the ß4-ß5 loops of YTH1 and YTH2 are distinct in length and amino acid composition. Our data revealed that, unlike in YTH1, the structure of m6A-binding pocket of YTH2 formed by the ß4-ß5 loop is stabilized by electrostatic interaction. This assignment and the structural information for YTH2 will provide the insight on the further functional research of YTHDC2.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Adenosine , RNA
7.
Macromol Biosci ; 20(10): e2000085, 2020 10.
Article in English | MEDLINE | ID: mdl-32734673

ABSTRACT

Hydrogels, whose degradability can be controlled while also preserving cell viability or biomolecule stability, are in demand. Degradable polyethylene glycol crosslinkers are hydrolytically designed for use in hydrogels. Degradation is controlled by crosslinker chemical structure, such as introducing local hydrophobicity, steric hindrance, or electron-withdrawing moieties near a degradable ester moiety. Hydrogels made using these crosslinkers have gelation times from 1 to 22 min, storage moduli from 3 to 10 kPa, mesh sizes from 10 to 13 nm, and degradation times from 18 h to 16 d. However, when reaction conditions are modified to achieve similar gelation time, hydrogels have similar initial properties but preserve the wide range of degradation times. All crosslinkers support high cell viability upon hydrogel encapsulation or exposure to leachables and degradation products. This innovation in controlling degradation can help realize the hydrogels' potential for drug delivery or as matrices for cell encapsulation and transplantation.


Subject(s)
Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Cell Line , Cell Survival , Elastic Modulus , Humans , Hydrolysis , Sulfhydryl Compounds/chemistry , Time Factors
8.
Antioxidants (Basel) ; 9(6)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599765

ABSTRACT

Myoglobin (Mb), an oxygen-binding heme protein highly expressed in heart and skeletal muscle, has been shown to undergo oxidative modifications on both an inter- and intramolecular level when exposed to hydrogen peroxide (H2O2) in vitro. Here, we show that exposure to H2O2 increases the peroxidase activity of Mb. Reaction of Mb with H2O2 causes covalent binding of heme to the Mb protein (Mb-X), corresponding to an increase in peroxidase activity when ascorbic acid is the reducing co-substrate. Treatment of H2O2-reacted Mb with ascorbic acid reverses the Mb-X crosslink. Reaction with H2O2 causes Mb to form dimers, trimers, and larger molecular weight Mb aggregates, and treatment with ascorbic acid regenerates Mb monomers. Reaction of Mb with H2O2 causes formation of dityrosine crosslinks, though the labile nature of the crosslinks broken by treatment with ascorbic acid suggests that the reversible aggregation of Mb is mediated by crosslinks other than dityrosine. Disappearance of a peptide containing a tryptophan residue when Mb is treated with H2O2 and the peptide's reappearance after subsequent treatment with ascorbic acid suggest that tryptophan side chains might participate in the labile crosslinking. Taken together, these data suggest that while exposure to H2O2 causes Mb-X formation, increases Mb peroxidase activity, and causes Mb aggregation, these oxidative modifications are reversible by treatment with ascorbic acid. A caveat is that future studies should demonstrate that these and other in vitro findings regarding properties of Mb have relevance in the intracellular milieu, especially in regard to actual concentrations of metMb, H2O2, and ascorbate that would be found in vivo.

9.
Structure ; 14(11): 1677-89, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17098193

ABSTRACT

The SF3a complex, consisting of SF3a60, SF3a66, and SF3a120, in 17S U2 snRNP is crucial to spliceosomal assembly. SF3a120 contains two tandem SURP domains (SURP1 and SURP2), and SURP2 is responsible for binding to SF3a60. We found that the SURP2 fragment forms a stable complex with an SF3a60 fragment (residues 71-107) and solved its structure by NMR spectroscopy. SURP2 exhibits a fold of the alpha1-alpha2-3(10)-alpha3 topology, and the SF3a60 fragment forms an amphipathic alpha helix intimately contacting alpha1 of SURP2. We also solved the SURP1 structure, which has the same fold as SURP2. The protein-binding interface of SURP2 is quite similar to the corresponding surface of SURP1, except for two amino acid residues. One of them, Leu169, is characteristic of SF3a120 SURP2 among SURP domains. Mutagenesis showed that this single Leu residue is the critical determinant for complex formation, which reveals the protein recognition mechanism in the subunit assembly.


Subject(s)
Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Amino Acid Sequence , Amino Acids/chemistry , DNA Mutational Analysis , Escherichia coli/metabolism , Humans , Leucine/chemistry , Magnetic Resonance Spectroscopy/methods , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid
10.
Nat Commun ; 9(1): 4229, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30315240

ABSTRACT

G-quadruplexes (G4s) are noncanonical DNA structures that frequently occur in the promoter regions of oncogenes, such as MYC, and regulate gene expression. Although G4s are attractive therapeutic targets, ligands capable of discriminating between different G4 structures are rare. Here, we describe DC-34, a small molecule that potently downregulates MYC transcription in cancer cells by a G4-dependent mechanism. Inhibition by DC-34 is significantly greater for MYC than other G4-driven genes. We use chemical, biophysical, biological, and structural studies to demonstrate a molecular rationale for the recognition of the MYC G4. We solve the structure of the MYC G4 in complex with DC-34 by NMR spectroscopy and illustrate specific contacts responsible for affinity and selectivity. Modification of DC-34 reveals features required for G4 affinity, biological activity, and validates the derived NMR structure. This work advances the design of quadruplex-interacting small molecules to control gene expression in therapeutic areas such as cancer.


Subject(s)
Cell Survival/physiology , G-Quadruplexes , Proto-Oncogene Proteins c-myc/metabolism , Binding Sites/genetics , Binding Sites/physiology , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Molecular Structure , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Protein Binding/genetics , Protein Binding/physiology , Protein Structure, Secondary , Proto-Oncogene Proteins c-myc/genetics , Structure-Activity Relationship , Surface Plasmon Resonance
11.
Protein Sci ; 16(8): 1577-87, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17656577

ABSTRACT

The zinc finger HIT domain is a sequence motif found in many proteins, including thyroid hormone receptor interacting protein 3 (TRIP-3), which is possibly involved in maturity-onset diabetes of the young (MODY). Novel zinc finger motifs are suggested to play important roles in gene regulation and chromatin remodeling. Here, we determined the high-resolution solution structure of the zinc finger HIT domain in ZNHIT2 (protein FON) from Homo sapiens, by an NMR method based on 567 upper distance limits derived from NOE intensities measured in three-dimensional NOESY spectra. The structure yielded a backbone RMSD to the mean coordinates of 0.19 A for the structured residues 12-48. The fold consists of two consecutive antiparallel beta-sheets and two short C-terminal helices packed against the second beta-sheet, and binds two zinc ions. Both zinc ions are coordinated tetrahedrally via a CCCC-CCHC motif to the ligand residues of the zf-HIT domain in an interleaved manner. The tertiary structure of the zinc finger HIT domain closely resembles the folds of the B-box, RING finger, and PHD domains with a cross-brace zinc coordination mode, but is distinct from them. The unique three-dimensional structure of the zinc finger HIT domain revealed a novel zinc-binding fold, as a new member of the treble clef domain family. On the basis of the structural data, we discuss the possible functional roles of the zinc finger HIT domain.


Subject(s)
Phosphoproteins/chemistry , Zinc Fingers , Amino Acid Sequence , Consensus Sequence , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphoproteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Solutions/chemistry , Surface Properties , Zinc/chemistry , Zinc/metabolism
13.
Cell Rep ; 14(11): 2683-94, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26971995

ABSTRACT

Myosin VI is critical for cargo trafficking and sorting during early endocytosis and autophagosome maturation, and abnormalities in these processes are linked to cancers, neurodegeneration, deafness, and hypertropic cardiomyopathy. We identify a structured domain in myosin VI, myosin VI ubiquitin-binding domain (MyUb), that binds to ubiquitin chains, especially those linked via K63, K11, and K29. Herein, we solve the solution structure of MyUb and MyUb:K63-linked diubiquitin. MyUb folds as a compact helix-turn-helix-like motif and nestles between the ubiquitins of K63-linked diubiquitin, interacting with distinct surfaces of each. A nine-amino-acid extension at the C-terminal helix (Helix2) of MyUb is required for myosin VI interaction with endocytic and autophagic adaptors. Structure-guided mutations revealed that a functional MyUb is necessary for optineurin interaction. In addition, we found that an isoform-specific helix restricts MyUb binding to ubiquitin chains. This work provides fundamental insights into myosin VI interaction with ubiquitinated cargo and functional adaptors.


Subject(s)
Myosin Heavy Chains/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Fluorescence Polarization , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , Ubiquitin/chemistry , Ubiquitin/genetics
14.
Nat Struct Mol Biol ; 23(4): 300-308, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950368

ABSTRACT

Myosin VI functions in endocytosis and cell motility. Alternative splicing of myosin VI mRNA generates two distinct isoform types, myosin VI(short) and myosin VI(long), which differ in the C-terminal region. Their physiological and pathological roles remain unknown. Here we identified an isoform-specific regulatory helix, named the α2-linker, that defines specific conformations and hence determines the target selectivity of human myosin VI. The presence of the α2-linker structurally defines a new clathrin-binding domain that is unique to myosin VI(long) and masks the known RRL interaction motif. This finding is relevant to ovarian cancer, in which alternative myosin VI splicing is aberrantly regulated, and exon skipping dictates cell addiction to myosin VI(short) in tumor-cell migration. The RRL interactor optineurin contributes to this process by selectively binding myosin VI(short). Thus, the α2-linker acts like a molecular switch that assigns myosin VI to distinct endocytic (myosin VI(long)) or migratory (myosin VI(short)) functional roles.


Subject(s)
Myosin Heavy Chains/chemistry , Myosin Heavy Chains/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell Movement , Clathrin/metabolism , Female , Gene Knockout Techniques , Humans , Models, Molecular , Molecular Sequence Data , Myosin Heavy Chains/genetics , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Magnetic Resonance, Biomolecular , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Protein Interaction Maps , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary
15.
FEBS Open Bio ; 4: 689-703, 2014.
Article in English | MEDLINE | ID: mdl-25161877

ABSTRACT

Anaphase-promoting complex or cyclosome (APC/C) is a multisubunit ubiquitin ligase E3 that targets cell-cycle regulators. Cdc20 is required for full activation of APC/C in M phase, and mediates substrate recognition. In vertebrates, Emi2/Erp1/FBXO43 inhibits APC/C-Cdc20, and functions as a cytostatic factor that causes long-term M phase arrest of mature oocytes. In this study, we found that a fragment corresponding to the zinc-binding region (ZBR) domain of Emi2 inhibits cell-cycle progression, and impairs the association of Cdc20 with the APC/C core complex in HEK293T cells. Furthermore, we revealed that the ZBR fragment of Emi2 inhibits in vitro ubiquitin chain elongation catalyzed by the APC/C cullin-RING ligase module, the ANAPC2-ANAPC11 subcomplex, in combination with the ubiquitin chain-initiating E2, E2C/UBE2C/UbcH10. Structural analyses revealed that the Emi2 ZBR domain uses different faces for the two mechanisms. Thus, the double-faced ZBR domain of Emi2 antagonizes the APC/C function by inhibiting both the binding with the coactivator Cdc20 and ubiquitylation mediated by the cullin-RING ligase module and E2C. In addition, the tail region between the ZBR domain and the C-terminal RL residues [the post-ZBR (PZ) region] interacts with the cullin subunit, ANAPC2. In the case of the ZBR fragment of the somatic paralogue of Emi2, Emi1/FBXO5, these inhibitory activities against cell division and ubiquitylation were not observed. Finally, we identified two sets of key residues in the Emi2 ZBR domain that selectively exert each of the dual Emi2-specific modes of APC/C inhibition, by their mutation in the Emi2 ZBR domain and their transplantation into the Emi1 ZBR domain.

16.
Nat Struct Mol Biol ; 21(9): 778-86, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25132178

ABSTRACT

Tissue-specific alternative pre-mRNA splicing is often cooperatively regulated by multiple splicing factors, but the structural basis of cooperative RNA recognition is poorly understood. In Caenorhabditis elegans, ligand binding specificity of fibroblast growth factor receptors (FGFRs) is determined by mutually exclusive alternative splicing of the sole FGFR gene, egl-15. Here we determined the solution structure of a ternary complex of the RNA-recognition motif (RRM) domains from the RBFOX protein ASD-1, SUP-12 and their target RNA from egl-15. The two RRM domains cooperatively interact with the RNA by sandwiching a G base to form the stable complex. Multichromatic fluorescence splicing reporters confirmed the requirement of the G and the juxtaposition of the respective cis elements for effective splicing regulation in vivo. Moreover, we identified a new target for the heterologous complex through an element search, confirming the functional significance of the intermolecular coordination.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Alternative Splicing , Animals , Base Sequence , Caenorhabditis elegans/chemistry , Caenorhabditis elegans Proteins/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , RNA/chemistry , RNA-Binding Proteins/chemistry
17.
FEBS Lett ; 586(21): 3858-64, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23010590

ABSTRACT

The WWE domain is often identified in proteins associated with ubiquitination or poly-ADP-ribosylation. Structural information about WWE domains has been obtained for the ubiquitination-related proteins, such as Deltex and RNF146, but not yet for the poly-ADP-ribose polymerases (PARPs). Here we determined the solution structures of the WWE domains from PARP11 and PARP14, and compared them with that of the RNF146 WWE domain. NMR perturbation experiments revealed the specific differences in their ADP-ribose recognition modes that correlated with their individual biological activities. The present structural information sheds light on the ADP-ribose recognition modes by the PARP WWE domains.


Subject(s)
Adenosine Diphosphate Ribose/chemistry , Poly(ADP-ribose) Polymerases/chemical synthesis , Ubiquitin-Protein Ligases/chemical synthesis , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , Humans , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Poly(ADP-ribose) Polymerases/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Ubiquitin-Protein Ligases/chemistry
18.
Structure ; 18(9): 1127-39, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826339

ABSTRACT

The zinc finger CW (zf-CW) domain is a motif of about 60 residues that is frequently found in proteins involved in epigenetic regulation. Here, we determined the NMR solution structure of the zf-CW domain of the human zf-CW and PWWP domain containing protein 1 (ZCWPW1). The zf-CW domain adopts a new fold in which a zinc ion is coordinated tetrahedrally by four conserved Cys ligand residues. The tertiary structure of the zf-CW domain partially resembles that adopted by the plant homeo domain (PHD) finger bound to the histone tail, suggesting that the zf-CW domain and the PHD finger have similar functions. The solution structure of the complex of the zf-CW domain with the histone H3 tail peptide (1-10) with trimethylated K4 clarified its binding mode. Our structural and biochemical studies have identified the zf-CW domain as a member of the histone modification reader modules for epigenetic regulation.


Subject(s)
Histones/chemistry , Zinc Fingers , Amino Acid Sequence , Binding Sites , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
19.
Protein Sci ; 18(3): 650-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19241374

ABSTRACT

Fn14 is the smallest member of the tumor necrosis factor (TNF) receptor superfamily, and specifically binds to its ligand, TWEAK (TNF-like weak inducer of apoptosis), which is a member of the TNF superfamily. The receptor-ligand recognition between Fn14 and TWEAK induces a variety of cellular processes for tissue remodeling and is also involved in the pathogenesis of some human diseases, such as cancer, chronic autoimmune diseases, and acute ischaemic stroke. The extracellular ligand-binding region of Fn14 is composed of 53 amino acid residues and forms a single, cysteine-rich domain (CRD). In this study, we determined the solution structure of the Fn14 CRD (Glu28-Ala70) by heteronuclear NMR, with a (13)C-/(15)N-labeled sample. The tertiary structure of the CRD comprises a beta-sheet with two strands, followed by a 3(10) helix and a C-terminal alpha-helix, and is stabilized by three disulfide bonds connecting Cys36-Cys49, Cys52-Cys67, and Cys55-Cys64. Comparison of the disulfide bond connectivities and the tertiary structures with those of other CRDs revealed that the Fn14 CRD is similar to the fourth CRD of TNF receptor 1 (A1-C2 module type), but not to the CRD of B-cell maturation antigen and the second CRD of transmembrane activator and CAML (calcium modulator and cyclophilin ligand) interactor (A1-D2 module type). This is the first structural report about the A1-C2 type CRD that could bind to the known target.


Subject(s)
Cysteine/chemistry , Receptors, Tumor Necrosis Factor/chemistry , Amino Acid Sequence , Cysteine/metabolism , Data Interpretation, Statistical , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor
20.
J Mol Biol ; 393(2): 478-95, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19683535

ABSTRACT

The Notch signaling pathway is critical for many developmental processes and requires complex trafficking of both Notch receptor and its ligands, Delta and Serrate. In Drosophila melanogaster, the endocytosis of Delta in the signal-sending cell is essential for Notch receptor activation. The Neuralized protein from D. melanogaster (Neur) is a ubiquitin E3 ligase, which binds to Delta through its first neuralized homology repeat 1 (NHR1) domain and mediates the ubiquitination of Delta for endocytosis. Tom, a Bearded protein family member, inhibits the Neur-mediated endocytosis through interactions with the NHR1 domain. We have identified the domain boundaries of the novel NHR1 domain, using a screening system based on our cell-free protein synthesis method, and demonstrated that the identified Neur NHR1 domain had binding activity to the 20-residue peptide corresponding to motif 2 of Tom by isothermal titration calorimetry experiments. We also determined the solution structure of the Neur NHR1 domain by heteronuclear NMR methods, using a (15)N/(13)C-labeled sample. The Neur NHR1 domain adopts a characteristic beta-sandwich fold, consisting of a concave five-stranded antiparallel beta-sheet and a convex seven-stranded antiparallel beta-sheet. The long loop (L6) between the beta6 and beta7 strands covers the hydrophobic patch on the concave beta-sheet surface, and the Neur NHR1 domain forms a compact globular fold. Intriguingly, in spite of the slight, but distinct, differences in the topology of the secondary structure elements, the structure of the Neur NHR1 domain is quite similar to those of the B30.2/SPRY domains, which are known to mediate specific protein-protein interactions. Further NMR titration experiments of the Neur NHR1 domain with the 20-residue Tom peptide revealed that the resonances originating from the bottom area of the beta-sandwich (the L3, L5, and L11 loops, as well as the tip of the L6 loop) were affected. In addition, a structural comparison of the Neur NHR1 domain with the first NHR domain of the human KIAA1787 protein, which is from another NHR subfamily and does not bind to the 20-residue Tom peptide, suggested the critical amino acid residues for the interactions between the Neur NHR1 domain and the Tom peptide. The present structural study will shed light on the role of the Neur NHR1 domain in the Notch signaling pathway.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/physiology , Drosophila melanogaster/enzymology , Receptors, Notch/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , Animals , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Magnetic Resonance Spectroscopy , Models, Biological , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction/genetics , Ubiquitin-Protein Ligases/classification , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL