Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Proteins ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38747678

ABSTRACT

FOXP2 is a transcription factor associated with speech and language. Like other FOX transcription factors, it has a DNA binding region called the forkhead domain (FHD). This domain can exist as a monomer or a domain swapped dimer. In addition to the FHD, the leucine zipper region (LZ) of FOXP2 is also believed to be associated with both DNA binding and oligomerization. To better understand the relationship between DNA binding and oligomerization of FOXP2, we investigated its structure, stability and dynamics, focusing specifically on the FHD and the LZ. We did this by using two constructs: one containing the isolated FHD and one containing both the LZ and the FHD (LZ-END). We demonstrate in this work, that while the FHD maintains a monomeric form that is capable of binding DNA, the LZ-END undergoes a dynamic transition between oligomeric states in the presence of DNA. Our findings suggest that FOXP2's LZ domain influences DNA binding affinity through a change in oligomeric state. We show through hydrogen exchange mass spectroscopy that certain parts of the FHD and interlinking region become less dynamic when in the presence of DNA, confirming DNA binding and oligomerization in these regions. Moreover, the detection of a stable equilibrium intermediate state during LZ-END unfolding supports the idea of cooperation between these two domains. Overall, our study sheds light on the interplay between two FOXP2 domains, providing insight into the protein's ability to respond dynamically to DNA, and enriching our understanding of FOXP2's role in gene regulation.

2.
Biochimie ; 221: 65-74, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38296156

ABSTRACT

Forkhead box P2 (FOXP2) regulates expression of various genes and is associated with language, speech and neural development as well as cancer. Since there may be a putative link between sex and language and because transcription factors rarely function in isolation, this study aims to investigate whether FOXP2 directly associates with oestrogen receptor α (ER1), a nuclear receptor responsible for sexual differentiation that is also associated with cancer. Isothermal titration calorimetry and fluorescence anisotropy were used to investigate the interaction between the DNA-binding forkhead domain (FHD) of FOXP2, the N-terminal region (NT) of FOXP2, and the ligand-binding domain (LBD) of ER1. ER1 LBD does not interact with FOXP2 NT but associates with apo-FOXP2 FHD in an enthalpically favourable manner. The affinity of this interaction is inversely correlated to the salt concentration. Additionally, FOXP2 FHD that is bound to ER1 LBD, has reduced ability to interact with its cognate DNA. This research identifies a novel interaction between ER1 LBD and FOXP2 FHD and shows that the interaction is regulated by salt. Moreover, FOXP2 FHD cannot bind to both ER1 LBD and DNA simultaneously, suggesting that this interaction could be involved in regulating the transcriptional pathway of FOXP2 should the interaction be found in vivo. This study could serve as a foundation for uncovering the basis of sexual dimorphism in speech and language development and related disorders and potentially offers an alternate for targeted cancer therapies.


Subject(s)
Estrogen Receptor alpha , Forkhead Transcription Factors , Protein Binding , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Humans , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/chemistry , Protein Domains , DNA/metabolism
3.
Biosci Rep ; 43(10)2023 10 31.
Article in English | MEDLINE | ID: mdl-37815922

ABSTRACT

YY1 is a ubiquitously expressed, intrinsically disordered transcription factor involved in neural development. The oligomeric state of YY1 varies depending on the environment. These structural changes may alter its DNA binding ability and hence its transcriptional activity. Just as YY1's oligomeric state can impact its role in transcription, so does its interaction with other proteins such as FOXP2. The aim of this work is to study the structure and dynamics of YY1 so as to determine the influence of oligomerisation and associations with FOXP2 on its DNA binding mechanism. The results confirm that YY1 is primarily a disordered protein, but it does consist of certain specific structured regions. We observed that YY1 quaternary structure is a heterogenous mixture of oligomers, the overall size of which is dependent on ionic strength. Both YY1 oligomerisation and its dynamic behaviour are further subject to changes upon DNA binding, whereby increases in DNA concentration result in a decrease in the size of YY1 oligomers. YY1 and the FOXP2 forkhead domain were found to interact with each other both in isolation and in the presence of YY1-specific DNA. The heterogeneous, dynamic multimerisation of YY1 identified in this work is, therefore likely to be important for its ability to make heterologous associations with other proteins such as FOXP2. The interactions that YY1 makes with itself, FOXP2 and DNA form part of an intricate mechanism of transcriptional regulation by YY1, which is vital for appropriate neural development.


Subject(s)
Intrinsically Disordered Proteins , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , DNA/metabolism , Gene Expression Regulation
4.
Molecules ; 28(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36985777

ABSTRACT

The unusual and interesting architecture of the catalytic chamber of the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) was recently explored using Cryogenic Electron Microscopy (Cryo-EM), which revealed the presence of two distinctive binding cavities within the catalytic chamber. In this report, first, we mapped out and fully characterized the variations between the two binding sites, BS1 and BS2, for significant differences in their amino acid architecture, size, volume, and hydrophobicity. This was followed by investigating the preferential binding of eight antiviral agents to each of the two binding sites, BS1 and BS2, to understand the fundamental factors that govern the preferential binding of each drug to each binding site. Results showed that, in general, hydrophobic drugs, such as remdesivir and sofosbuvir, bind better to both binding sites than relatively less hydrophobic drugs, such as alovudine, molnupiravir, zidovudine, favilavir, and ribavirin. However, suramin, which is a highly hydrophobic drug, unexpectedly showed overall weaker binding affinities in both binding sites when compared to other drugs. This unexpected observation may be attributed to its high binding solvation energy, which disfavors overall binding of suramin in both binding sites. On the other hand, hydrophobic drugs displayed higher binding affinities towards BS1 due to its higher hydrophobic architecture when compared to BS2, while less hydrophobic drugs did not show a significant difference in binding affinities in both binding sites. Analysis of binding energy contributions revealed that the most favorable components are the ΔEele, ΔEvdw, and ΔGgas, whereas ΔGsol was unfavorable. The ΔEele and ΔGgas for hydrophobic drugs were enough to balance the unfavorable ΔGsol, leaving the ΔEvdw to be the most determining factor of the total binding energy. The information presented in this report will provide guidelines for tailoring SARS-CoV-2 inhibitors with enhanced binding profiles.


Subject(s)
COVID-19 , Humans , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/metabolism , RNA, Viral , Suramin , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/chemistry , Molecular Docking Simulation
5.
Biosci Rep ; 41(1)2021 01 29.
Article in English | MEDLINE | ID: mdl-33319247

ABSTRACT

FOX proteins are a superfamily of transcription factors which share a DNA-binding domain referred to as the forkhead domain. Our focus is on the FOXP subfamily members, which are involved in language and cognition amongst other things. The FOXP proteins contain a conserved zinc finger and a leucine zipper motif in addition to the forkhead domain. The remainder of the sequence is predicted to be unstructured and includes an acidic C-terminal tail. In the present study, we aim to investigate how both the structured and unstructured regions of the sequence cooperate so as to enable FOXP proteins to perform their function. We do this by studying the effect of these regions on both oligomerisation and DNA binding. Structurally, the FOXP proteins appear to be comparatively globular with a high proportion of helical structure. The proteins multimerise via the leucine zipper, and the stability of the multimers is controlled by the unstructured interlinking sequence including the acid rich tail. FOXP2 is more compact than FOXP1, has a greater propensity to form higher order oligomers, and binds DNA with stronger affinity. We conclude that while the forkhead domain is necessary for DNA binding, the affinity of the binding event is attributable to the leucine zipper, and the unstructured regions play a significant role in the specificity of binding. The acid rich tail forms specific contacts with the forkhead domain which may influence oligomerisation and DNA binding, and therefore the acid rich tail may play an important regulatory role in FOXP transcription.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Biopolymers/chemistry , Biopolymers/metabolism , Chromatography, Gel , Circular Dichroism , Leucine Zippers , Protein Binding , Protein Domains , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
6.
Mol Biochem Parasitol ; 240: 111319, 2020 11.
Article in English | MEDLINE | ID: mdl-32961204

ABSTRACT

Schistosoma japonicum glutathione transferase (Sj26GST), an enzyme central to detoxification of electrophilic compounds in the parasite, is upregulated in response to drug treatment. Therefore, Sj26GST may serve as a potential therapeutic target for the treatment of schistosomiasis. Herewith, we describe the structural basis of inhibition of Sj26GST by ellagic acid (EA). Using 1-chloro-2,4-dinitrobenzene and reduced glutathione (GSH) as Sj26GST substrates, EA was shown to inhibit Sj26GST activity by 66 % with an IC50 of 2.4 µM. Fluorescence spectroscopy showed that EA altered the polarity of the environment of intrinsic tryptophan and that EA decreased (in a dose-dependent manner) the interaction between Sj26GST and 8-Anilino-1-naphthalenesulfonate (ANS), which is a known GST H-site ligand. Thermodynamic studies indicated that the interaction between Sj26GST and EA is spontaneous (ΔG = -29.88 ± 0.07 kJ/mol), enthalpically-driven (ΔH = -9.48 ± 0.42 kJ/mol) with a favourable entropic change (ΔS = 20.40 ± 0.08 kJ/mol/K), and with a stoichiometry of four EA molecules bound per Sj26GST dimer. The 1.53 Å-resolution Sj26GST crystal structure (P 21 21 21 space group) complexed with GSH and EA shows that EA binds primarily at the dimer interface, stabilised largely by Van der Waal forces and H-bonding. Besides, EA bound near the H-site and less than 3.5 Å from the ε-NH2 of the γ-glutamyl moiety of GSH, in each subunit.


Subject(s)
Enzyme Inhibitors/chemistry , Glutathione Transferase/chemistry , Helminth Proteins/chemistry , Schistosoma japonicum/enzymology , Animals , Calorimetry , Chemical Phenomena , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Helminth Proteins/antagonists & inhibitors , Kinetics , Ligands , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Schistosoma japonicum/drug effects , Structure-Activity Relationship , Thermodynamics
7.
Biochemistry ; 59(36): 3332-3346, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786407

ABSTRACT

H1.2 is a key mediator of apoptosis following DNA double-strand breaks. The link between H1.2 and canonical apoptotic pathways is unclear. One study found that H1.2 stimulates cytochrome c (Cyt c) release; in contrast, apoptosis-inducing factor was found to be released in another study. The C-terminal domain (CTD) of H1.2 has been implicated in the latter pathway, but activation of the proapoptotic protein BCL-2 homologous antagonist/killer (BAK) is a common denominator in both pathways. This study aimed to determine whether the CTD of H1.2 is also responsible for mitochondrial Cyt c release and whether a previously identified K/RVVKP motif in the CTD mediates the response. This study investigated if H1.2 mediates apoptosis induction through direct interaction with BAK. We established that the CTD of H1.2 stimulates mitochondrial Cyt c release in vitro in a mitochondrial permeability transition-independent manner and that the substitution of a single valine with threonine in the K/RVVKP motif abolishes Cyt c release. Additionally, we showed that H1.2 directly interacts with BAK with weak affinity and that the CTD of H1.2 mediates this binding. Using two 20-amino acid peptides derived from the CTD of H1.2 and H1.1 (K/RVVKP motif inclusive), we determined the main residues involved in the direct interaction with BAK. We propose that H1.2 operates through the K/RVVKP motif by directly activating BAK through inter- and intramolecular interactions. These findings expand the view of H1.2 as a signal-transducing molecule that can activate apoptosis in a BAK-dependent manner.


Subject(s)
Apoptosis , Cytochromes c/metabolism , Histones/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Amino Acid Motifs , Amino Acid Sequence , Histones/chemistry , Humans , Mitochondria/metabolism , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Protein Domains , Sequence Homology , bcl-2 Homologous Antagonist-Killer Protein/chemistry
8.
Arch Biochem Biophys ; 657: 56-64, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30227110

ABSTRACT

FOXP2 is a transcriptional repressor involved in development of the human brain and is the first gene product to be linked to the evolution of human speech. FOXP2 belongs to the FOX superfamily of proteins that share a common winged helix DNA binding domain - the forkhead domain. A divalent cation (Mg2+ or Ca2+) has been identified bound to a group of highly conserved residues in a number of FOX forkhead domain crystal structures. This work aims to investigate the role of the conserved divalent cation binding site by studying both the structure and DNA-binding function of the FOXP2 forkhead domain when in the presence and absence of either cation (Mg2+or Ca2+). The presence of the cations does not significantly alter the structure of the apo-FOXP2 forkhead domain. However, when in the presence of a cognate oligonucleotide sequence, differences are observed upon addition of divalent cation. These differences occur both in the structure and in the thermodynamic DNA binding signature of the FOXP2 forkhead domain. The incorporation of molecular dynamics simulations together with the experimental data provides us with sufficient insight so as to propose a possible role for divalent cations in the regulation of DNA binding to FOX transcription factors.


Subject(s)
Calcium/metabolism , DNA/metabolism , Forkhead Transcription Factors/metabolism , Magnesium/metabolism , Amino Acid Sequence , Binding Sites , Escherichia coli/genetics , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protein Structure, Tertiary , Sequence Alignment , Thermodynamics
9.
Biol Chem ; 399(8): 881-893, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29878882

ABSTRACT

Forkhead box (FOX) proteins are a ubiquitously expressed family of transcription factors that regulate the development and differentiation of a wide range of tissues in animals. The FOXP subfamily members are the only known FOX proteins capable of forming domain-swapped forkhead domain (FHD) dimers. This is proposed to be due to an evolutionary mutation (P539A) that lies in the FHD hinge loop, a key region thought to fine-tune DNA sequence specificity in the FOX transcription factors. Considering the importance of the hinge loop in both the dimerisation mechanism of the FOXP FHD and its role in tuning DNA binding, a detailed investigation into the implications of mutations within this region could provide important insight into the evolution of the FOX family. Isothermal titration calorimetry and hydrogen exchange mass spectroscopy were used to study the thermodynamic binding signature and changes in backbone dynamics of FOXP2 FHD DNA binding. Dual luciferase reporter assays were performed to study the effect that the hinge-loop mutation has on FOXP2 transcriptional activity in vivo. We demonstrate that the change in dynamics of the hinge-loop region of FOXP2 alters the energetics and mechanism of DNA binding highlighting the critical role of hinge loop mutations in regulating DNA binding characteristics of the FOX proteins.


Subject(s)
DNA/chemistry , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Transcription, Genetic , Binding Sites , DNA/metabolism , Humans
10.
Protein J ; 37(4): 311-323, 2018 08.
Article in English | MEDLINE | ID: mdl-29845391

ABSTRACT

FOXP2 is a transcription factor expressed in multiple tissues during embryonic development. FOXP2 regulates transcription by binding to DNA at its DNA binding domain, the forkhead domain (FHD) through the recognition helix. Ser557 is a residue located within the recognition helix that has the potential to become phosphorylated posttranslationally. In this study we investigated whether phosphorylation of Ser557 can influence the structure and DNA binding of the FOXP2 FHD. We did this by constructing S557E, a phosphomimetic mutant, and comparing its behaviour to the wild type. The mutation did not affect the secondary or tertiary structure of the protein although it did decrease the propensity of the FOXP2 FHD to form dimers. Most notably, the mutation showed significantly reduced DNA binding compared to the wild type as detected using electrophoretic mobility shift assays. Molecular docking was also performed in which the wild type, phosphomimetic mutant and phosphorylated wild-type were docked to DNA and their interactions with DNA were compared. These results indicated that the wild type forms more interactions with the DNA and that the phosphomimetic mutant as well as the phosphorylated wild type did not associate as favourably with the DNA. This indicates that phosphorylation of Ser557 could disrupt DNA binding likely due to electrostatic and steric hindrance. This suggests that phosphorylation of Ser557 in the FOXP2 FHD could act as a control mechanism for FOXP2 and ultimately could be involved in regulation of transcription.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/metabolism , Mutation , Serine/metabolism , DNA/chemistry , DNA/genetics , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , Humans , Molecular Docking Simulation , Molecular Mimicry , Phosphorylation , Protein Conformation , Protein Domains , Serine/chemistry , Serine/genetics
11.
J Biochem ; 162(1): 45-54, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28104810

ABSTRACT

FOXP2 is a member of the P subfamily of FOX transcription factors, the DNA-binding domain of which is the winged helix forkhead domain (FHD). In this work we show that the FOXP2 FHD is able to bind to various DNA sequences, including a novel sequence identified in this work, with different affinities and rates as detected using surface plasmon resonance. Combining the experimental work with molecular docking, we show that high-affinity sequences remain bound to the protein for longer, form a greater number of interactions with the protein and induce a greater structural change in the protein than low-affinity sequences. We propose a binding model for the FOXP2 FHD that involves three types of binding sequence: low affinity sites which allow for rapid scanning of the genome by the protein in a partially unstructured state; moderate affinity sites which serve to locate the protein near target sites and high-affinity sites which secure the protein to the DNA and induce a conformational change necessary for functional binding and the possible initiation of downstream transcriptional events.


Subject(s)
DNA/genetics , DNA/metabolism , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Base Sequence , Binding Sites , Humans , Models, Molecular , Protein Domains , Surface Plasmon Resonance
12.
Biochemistry ; 55(13): 1959-67, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26950495

ABSTRACT

Forkhead box (FOX) transcription factors share a conserved forkhead DNA binding domain (FHD) and are key role players in the development of many eukaryotic species. Their involvement in various congenital disorders and cancers makes them clinically relevant targets for novel therapeutic strategies. Among them, the FOXP subfamily of multidomain transcriptional repressors is unique in its ability to form DNA binding homo and heterodimers. The truncated FOXP2 FHD, in the absence of the leucine zipper, exists in equilibrium between monomeric and domain-swapped dimeric states in vitro. As a consequence, determining the DNA binding properties of the FOXP2 FHD becomes inherently difficult. In this work, two FOXP2 FHD hinge loop mutants have been generated to successfully prevent both the formation (A539P) and the dissociation (F541C) of the homodimers. This allows for the separation of the two species for downstream DNA binding studies. Comparison of DNA binding of the different species using electrophoretic mobility shift assay, fluorescence anisotropy and isothermal titration calorimetry indicates that the wild-type FOXP2 FHD binds DNA as a monomer. However, comparison of the DNA-binding energetics of the monomer and wild-type FHD, reveals that there is a difference in the mechanism of binding between the two species. We conclude that the naturally occurring reverse mutation (P539A) seen in the FOXP subfamily increases DNA binding affinity and may increase the potential for nonspecific binding compared to other FOX family members.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/metabolism , Models, Molecular , Mutant Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Conserved Sequence , DNA/chemistry , Dimerization , Electrophoretic Mobility Shift Assay , Evolution, Molecular , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/genetics , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment
13.
Biochemistry ; 54(25): 4001-7, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26055196

ABSTRACT

Forkhead box P2 (FOXP2) is a transcription factor expressed in cardiovascular, intestinal, and neural tissues during embryonic development and is implicated in language development. FOXP2 like other FOX proteins contains a DNA binding domain known as the forkhead domain (FHD). The FHD interacts with DNA by inserting helix 3 into the major groove. One of these DNA-protein interactions is a direct hydrogen bond that is formed with His554. FOXP2 is localized in the nuclear compartment that has a pH of 7.5. Histidine contains an imidazole side chain in which the amino group typically has a pKa of ~6.5. It seems possible that pH fluctuations around 6.5 may result in changes in the protonation state of His554 and thus the ability of the FOXP2 FHD to bind DNA. To investigate the effect of pH on the FHD, both the structure and the binding affinity were studied in the pH range of 5-9. This was done in the presence and absence of DNA. The structure was assessed using size exclusion chromatography, far-UV circular dichroism, and intrinsic and extrinsic fluorescence. The results indicated that while pH did not affect the secondary structure in the presence or absence of DNA, the tertiary structure was pH sensitive and the protein was less compact at low pH. Furthermore, the presence of DNA caused the protein to become more compact at low pH and also had the potential to increase the dimerization propensity. Fluorescence anisotropy was used to investigate the effect of pH on the FOXP2 FHD DNA binding affinity. It was found that pH had a direct effect on binding affinity. This was attributed to the altered hydrogen bonding patterns upon protonation or deprotonation of His554. These results could implicate pH as a means of regulating transcription by the FOXP2 FHD, which may also have repercussions for the behavior of this protein in cancer cells.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Amino Acid Sequence , DNA/chemistry , Forkhead Transcription Factors/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Protein Conformation , Protein Structure, Tertiary
14.
Protein J ; 34(2): 111-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724387

ABSTRACT

The forkhead box (FOX) proteins are a family of transcription factors that interact with DNA via a winged helix motif that forms part of the forkhead domain. The FOXP (FOXP1-4) subfamily is unique in the family in that the forkhead domains of these proteins are able to dimerise via domain swapping. In this event, structural elements are exchanged via extension of the hinge loop region. Despite the high sequence homology among the FOXP subfamily members, the stability of their forkhead domain dimers varies, with FOXP3 forming the most stable dimer. An amino acid difference is observed in the hinge region of the FOXP subfamily where a tyrosine in all members is replaced with a phenylalanine in FOXP3. In this work, the role of phenylalanine at this position in the hinge region was investigated. This was done by creating the Y540F variant of the FOXP2 forkhead domain. The effect of the Y540F mutation on the structure, dimerisation propensity and DNA binding ability of the FOXP subfamily was investigated. The mutation altered the structure of the protein by decreasing the disorder of the backbone as measured by circular dichroism spectroscopy and by altering the local environment of the hinge region as measured by tryptophan fluorescence. The propensity of the forkhead domain to form a dimer was improved ~9.5 fold by the mutation. This was attributed to increased hydrophobicity at the dimer interface as well as altered tension in the hinge loop region. DNA binding assays indicated that the affinity for DNA was decreased by the mutation. Taken together, these findings suggest that domain swapping may modulate DNA binding.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Forkhead Transcription Factors/chemistry , Protein Multimerization , Tyrosine/chemistry , Amino Acid Sequence , Chromatography, Gel , Circular Dichroism , Crystallography, X-Ray , DNA/genetics , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Forkhead Transcription Factors/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Alignment , Tyrosine/genetics
15.
Mol Cell Biochem ; 398(1-2): 83-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25209805

ABSTRACT

The chloride intracellular channel protein, CLIC1, is synthesised as a soluble monomer that can reversibly bind membranes. Soluble CLIC1 is proposed to respond to the low pH found at a membrane surface by partially unfolding and restructuring into a membrane-competent conformation. This transition is proposed to be controlled by strategically located "pH-sensor" residues that become protonated at acidic pH. In this study, we investigate the role of two conserved glutamate residues, Glu85 in the N-domain and Glu228 in the C-domain, as pH-sensors. E85L and E228L CLIC1 variants were created to reduce pH sensitivity by permanently breaking the bonds these residues form. The structure and stability of each variant was compared to the wild type at both pH 7.0 and pH 5.5. Neither substitution significantly altered the structure but both decreased the conformational stability. Furthermore, E85L CLIC1 formed a urea-induced unfolding intermediate state at both pH 7 and pH 5.5 compared to wild-type and E228L CLIC1 which only formed the intermediate at pH 5.5. We conclude that Glu85 and Glu228 are two of the five pH-sensor residues of CLIC1 and contribute to the pH-response in different ways. Glu228 lowers the stability of the native state at pH 5.5, while Glu85 contributes both to the stability of the native state and to the formation of the intermediate state. By putting these interactions into the context of the three previously described CLIC1 pH-sensor residues, we propose a mechanism for the conversion of CLIC1 from the soluble state to the pre-membrane form.


Subject(s)
Chloride Channels/chemistry , Glutamic Acid/chemistry , Membrane Proteins/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Chloride Channels/genetics , Circular Dichroism , Glutamic Acid/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation, Missense , Protein Stability , Protein Unfolding/drug effects , Sequence Homology, Amino Acid , Solubility , Spectrometry, Fluorescence , Temperature , Urea/pharmacology
16.
Eur Biophys J ; 43(8-9): 405-14, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24925575

ABSTRACT

The chloride intracellular channel protein 1 (CLIC1) is unique among eukaryotic ion channels in that it can exist as either a soluble monomer or an integral membrane channel. CLIC1 contains no known membrane-targeting signal sequences and the environmental factors which promote membrane binding of the transmembrane domain (TMD) are poorly understood. Here we report a positively charged motif at the C-terminus of the TMD and show that it enhances membrane partitioning and insertion. A 30-mer TMD peptide was synthesized in which the positively charged motif was replaced by three glutamate residues. The peptide was examined in 2,2,2-trifluoroethanol (TFE), sodium dodecyl sulfate micelles and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine liposomes using size-exclusion chromatography, far-UV CD, and fluorescence spectroscopy. The motif appears to enhance membrane interaction via electrostatic contacts and functions as an electrostatic plug to anchor the TMD in membranes. In addition, the motif is also involved in orientating the TMD with respect to the cis and trans faces of the membrane. These findings shed light on the intrinsic and environmental factors that promote the spontaneous conversion of CLIC1 from a water-soluble to a membrane-bound protein.


Subject(s)
Cell Membrane/metabolism , Chloride Channels/chemistry , Conserved Sequence , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Adsorption , Amino Acid Motifs , Amino Acid Sequence , Biological Transport , Cell Membrane/chemistry , Chlorides/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Solvents/chemistry
17.
Biochemistry ; 53(1): 57-67, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24328417

ABSTRACT

Chloride intracellular channel protein 1 (CLIC1) is a dual-state protein that can exist either as a soluble monomer or in an integral membrane form. The oligomerization of the transmembrane domain (TMD) remains speculative despite it being implicated in pore formation. The extent to which electrostatic and van der Waals interactions drive folding and association of the dimorphic TMD is unknown and is complicated by the requirement of interactions favorable in both aqueous and membrane environments. Here we report a putative Lys37-Trp35 cation-π interaction and show that it stabilizes the dimeric form of the CLIC1 TMD in membranes. A synthetic 30-mer peptide comprising a K37M TMD mutant was examined in 2,2,2-trifluoroethanol, sodium dodecyl sulfate micelles, and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine liposomes using far-ultraviolet (UV) circular dichroism, fluorescence, and UV absorbance spectroscopy. Our data suggest that Lys37 is not implicated in the folding, stability, or membrane insertion of the TMD peptide. However, removal of this residue impairs the formation of dimers and higher-order oligomers. This is accompanied by a 30-fold loss of chloride influx activity, suggesting that dimerization modulates the rate of chloride conductance. We propose that, within membranes, individual TMD helices associate via a Lys37-mediated cation-π interaction to form active dimers. The latter findings are also supported by results of modeling a putative TMD dimer conformation in which Lys37 and Trp35 form cation-π pairs at the dimer interface. Dimeric helix bundles may then associate to form fully active ion channels. Thus, within a membrane-like environment, aromatic interactions involving a polar lysine side chain provide a thermodynamic driving force for helix-helix association.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/physiology , Lysine/chemistry , Protein Multimerization/physiology , Tryptophan/chemistry , Amino Acid Sequence , Chloride Channels/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Peptide Fragments/chemistry , Protein Structure, Tertiary
18.
Biochemistry ; 52(16): 2739-49, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23547926

ABSTRACT

Chloride intracellular channel protein 1 (CLIC1) is a dual-state protein that can exist either as a soluble monomer or in an integral membrane form. The transmembrane domain (TMD), implicated in membrane penetration and pore formation, comprises helix α1 and strand ß2 of the N-domain of soluble CLIC1. The mechanism by which the TMD binds, inserts, and oligomerizes in membranes to form a functional chloride channel is unknown. Here we report the secondary, tertiary, and quaternary structural changes of the CLIC1 TMD as it partitions between an aqueous and membrane-mimicking environment. A synthetic 30-mer peptide comprising the TMD was examined in 2,2,2-trifluoroethanol, sodium dodecyl sulfate (SDS) micelles, and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) liposomes using far-ultraviolet circular dichroism and fluorescence spectroscopy. Data obtained in the presence of SDS micelles and POPC liposomes show that Trp35 and Cys24 have reduced solvent accessibility, indicating that the peptide adopts an inserted orientation. The peptide assumes a helical structure in the presence of these mimetics, consistent with its predicted membrane conformation. This acquisition of secondary structure is concentration-dependent, suggesting an oligomerization event. Stable dimeric and trimeric species were subsequently identified using SDS-polyacrylamide gel electrophoresis. We propose that, in the vicinity of membranes, the mixed α/ß TMD in CLIC1 rearranges to form a helix that then likely dimerizes via noncovalent helix-helix interactions to form a membrane-competent protopore complex. Such oligomerization would be essential for forming a functional ion channel, given that each CLIC1 monomer possesses only a single TMD. This work highlights the central role of the TMD in CLIC1 function: It is capable of promoting membrane insertion and dimerization in the absence of the C-domain and large portions of the N-domain.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/metabolism , Amino Acid Sequence , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Liposomes/chemistry , Models, Molecular , Molecular Sequence Data , Phosphatidylcholines/chemistry , Protein Conformation , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Fluorescence , Trifluoroethanol/chemistry
19.
J Biomol Struct Dyn ; 31(12): 1370-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23140382

ABSTRACT

The HIV protease plays a major role in the life cycle of the virus and has long been a target in antiviral therapy. Resistance of HIV protease to protease inhibitors (PIs) is problematic for the effective treatment of HIV infection. The South African HIV-1 subtype C protease (C-SA PR), which contains eight polymorphisms relative to the consensus HIV-1 subtype B protease, was expressed in Escherichia coli, purified, and crystallized. The crystal structure of the C-SA PR was resolved at 2.7 Å, which is the first crystal structure of a HIV-1 subtype C protease that predominates in Africa. Structural analyses of the C-SA PR in comparison to HIV-1 subtype B proteases indicated that polymorphisms at position 36 of the homodimeric HIV-1 protease may impact on the stability of the hinge region of the protease, and hence the dynamics of the flap region. Molecular dynamics simulations showed that the flap region of the C-SA PR displays a wider range of movements over time as compared to the subtype B proteases. Reduced stability in the hinge region resulting from the absent E35-R57 salt bridge in the C-SA PR, most likely contributes to the increased flexibility of the flaps which may be associated with reduced susceptibility to PIs.


Subject(s)
Drug Resistance, Viral/drug effects , HIV Protease Inhibitors/pharmacology , HIV Protease/chemistry , HIV-1/drug effects , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Drug Resistance, Viral/genetics , Enzyme Stability/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease/genetics , HIV Protease/metabolism , HIV Protease Inhibitors/therapeutic use , HIV-1/enzymology , HIV-1/genetics , Humans , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Polymorphism, Genetic , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , South Africa
20.
Biochemistry ; 51(40): 7854-62, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22966869

ABSTRACT

The ion channel protein CLIC1 exists in both a soluble conformation in the cytoplasm and a membrane-bound conformation. The conformational stability of soluble CLIC1 demonstrates pH sensitivity which may be attributable to very specific residues that function as pH sensors. These sensors could be histidine or glutamate residues with pK(a) values that fall within the physiological pH range. The role of Glu81, a member of a topologically conserved buried salt bridge in CLIC1, as a pH sensor was investigated here. The mutants E81M, R29M, and E81M/R29M were designed to break the salt bridge between Glu81 and Arg29 and examine the effect of each member on the stability of the protein. Spectroscopic studies and the solved crystal structures indicated that the global structure of CLIC1 was not affected by the mutations. Urea-induced equilibrium unfolding unexpectedly showed E81M to stabilize CLIC1 at pH 7. This was due to stabilizing hydrophobic interactions with Met81 and a water-mediated compensatory H-bond between Met81 and Arg29. R29M and E81M/R29M destabilized CLIC1 at pH 7, and the unfolding transition changed from two-state to three-state, mimicking the wild type at pH 5.5. This observation points out the significance of the salt bridge in stabilizing the native state. The total unfolding free energy change of E81M CLIC1 does not change with pH, implying that Glu81 forms one of a network of pH-sensor residues in CLIC1 responsible for destabilization of the native state. This allows detachment of the N-domain from the C-domain at low pH.


Subject(s)
Arginine/chemistry , Chloride Channels/chemistry , Glutamic Acid/chemistry , Amino Acid Sequence , Crystallization , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Folding , Protein Stability , Spectrometry, Fluorescence , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...