Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Am Chem Soc ; 143(40): 16401-16410, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34606279

ABSTRACT

Biomimetics is a design principle within chemistry, biology, and engineering, but chemistry biomimetic approaches have been generally limited to emulating nature's chemical toolkit while emulation of nature's physical toolkit has remained largely unexplored. To begin to explore this, we designed biophysically mimetic microfluidic reactors with characteristic length scales and shear stresses observed within capillaries. We modeled the effect of shear with molecular dynamics studies and showed that this induces specific normally buried residues to become solvent accessible. We then showed using kinetics experiments that rates of reaction of these specific residues in fact increase in a shear-dependent fashion. We applied our results in the creation of a new microfluidic approach for the multidimensional study of cysteine biomarkers. Finally, we used our approach to establish dissociation of the therapeutic antibody trastuzumab in a reducing environment. Our results have implications for the efficacy of existing therapeutic antibodies in blood plasma as well as suggesting in general that biophysically mimetic chemistry is exploited in biology and should be explored as a research area.


Subject(s)
Biomimetics
2.
Biochemistry ; 59(48): 4563-4572, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33237763

ABSTRACT

The initial state of the intrinsically disordered protein α-synuclein (aSyn), e.g., the presence of oligomers and degradation products, or the presence of contaminants and adducts can greatly influence the aggregation kinetics and toxicity of the protein. Here, we compare four commonly used protocols for the isolation of recombinant aSyn from Escherichia coli: boiling, acid precipitation, ammonium sulfate precipitation, and periplasmic lysis followed by ion exchange chromatography and gel filtration. We identified, using nondenaturing electrospray ionization mass spectrometry, that aSyn isolated by acid precipitation and periplasmic lysis was the purest and yielded the highest percentage of monomeric protein, 100% and 96.5%, respectively. We then show that aSyn purified by the different protocols exerts different metabolic stresses in cells, with the more multimeric/degraded and least pure samples leading to a larger increase in cell vitality. However, the percentage of monomeric protein and the purity of the samples did not correlate with aSyn aggregation propensity. This study highlights the importance of characterizing monomeric aSyn after purification, as the choice of purification method can significantly influence the outcome of a subsequent study.


Subject(s)
alpha-Synuclein/isolation & purification , Cell Line , Cell Survival , Chemical Precipitation , Chromatography, Gel , Chromatography, Ion Exchange , Chromatography, Liquid , Escherichia coli/chemistry , Escherichia coli/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/isolation & purification , Microscopy, Electron, Transmission , Protein Aggregates , Protein Conformation , Protein Conformation, beta-Strand , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Spectrometry, Mass, Electrospray Ionization , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
4.
BMC Biol ; 16(1): 76, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29996828

ABSTRACT

BACKGROUND: Protein quality control mechanisms are essential for cell health and involve delivery of proteins to specific cellular compartments for recycling or degradation. In particular, stray hydrophobic proteins are captured in the aqueous cytosol by a co-chaperone, the small glutamine-rich, tetratricopeptide repeat-containing protein alpha (SGTA), which facilitates the correct targeting of tail-anchored membrane proteins, as well as the sorting of membrane and secretory proteins that mislocalize to the cytosol and endoplasmic reticulum-associated degradation. Full-length SGTA has an unusual elongated dimeric structure that has, until now, evaded detailed structural analysis. The C-terminal region of SGTA plays a key role in binding a broad range of hydrophobic substrates, yet in contrast to the well-characterized N-terminal and TPR domains, there is a lack of structural information on the C-terminal domain. In this study, we present new insights into the conformation and organization of distinct domains of SGTA and show that the C-terminal domain possesses a conserved region essential for substrate processing in vivo. RESULTS: We show that the C-terminal domain region is characterized by α-helical propensity and an intrinsic ability to dimerize independently of the N-terminal domain. Based on the properties of different regions of SGTA that are revealed using cell biology, NMR, SAXS, Native MS, and EPR, we observe that its C-terminal domain can dimerize in the full-length protein and propose that this reflects a closed conformation of the substrate-binding domain. CONCLUSION: Our results provide novel insights into the structural complexity of SGTA and provide a new basis for mechanistic studies of substrate binding and release at the C-terminal region.


Subject(s)
Carrier Proteins/chemistry , Molecular Chaperones/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Protein Binding , Protein Domains , Protein Multimerization , Protein Transport , Scattering, Small Angle
5.
Nucleic Acids Res ; 46(1): 387-402, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29136196

ABSTRACT

The RNA degradosome is a multi-enzyme assembly that plays a central role in the RNA metabolism of Escherichia coli and numerous other bacterial species including pathogens. At the core of the assembly is the endoribonuclease RNase E, one of the largest E. coli proteins and also one that bears the greatest region predicted to be natively unstructured. This extensive unstructured region, situated in the C-terminal half of RNase E, is punctuated with conserved short linear motifs that recruit partner proteins, direct RNA interactions, and enable association with the cytoplasmic membrane. We have structurally characterized a subassembly of the degradosome-comprising a 248-residue segment of the natively unstructured part of RNase E, the DEAD-box helicase RhlB and the glycolytic enzyme enolase, and provide evidence that it serves as a flexible recognition centre that can co-recruit small regulatory RNA and the RNA chaperone Hfq. Our results support a model in which the degradosome captures substrates and regulatory RNAs through the recognition centre, facilitates pairing to cognate transcripts and presents the target to the ribonuclease active sites of the greater assembly for cooperative degradation or processing.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/metabolism , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , RNA, Bacterial/metabolism , Binding Sites/genetics , Crystallography, X-Ray , Endoribonucleases/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , Models, Molecular , Multienzyme Complexes/genetics , Nucleic Acid Conformation , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Binding , Protein Domains , RNA Helicases/genetics , RNA, Bacterial/chemistry , RNA, Bacterial/genetics
6.
J Nanobiotechnology ; 15(1): 70, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-28985740

ABSTRACT

BACKGROUND: Due to their natural tendency to self-assemble, proteins and peptides are important components for organic nanotechnology. One particular class of peptides of recent interest is those that form amyloid fibrils, as this self-assembly results in extremely strong, stable quasi-one-dimensional structures which can be used to organise a wide range of cargo species including proteins and oligonucleotides. However, assembly of peptides already conjugated to proteins is limited to cargo species that do not interfere sterically with the assembly process or misfold under the harsh conditions often used for assembly. Therefore, a general method is needed to conjugate proteins and other molecules to amyloid fibrils after the fibrils have self-assembled. RESULTS: Here we have designed an amyloidogenic peptide based on the TTR105-115 fragment of transthyretin to form fibrils that display an alkyne functionality, important for bioorthogonal chemical reactions, on their surface. The fibrils were formed and reacted both with an azide-containing amino acid and with an azide-functionalised dye by the Huisgen cycloaddition, one of the class of "click" reactions. Mass spectrometry and total internal reflection fluorescence optical microscopy were used to show that peptides incorporated into the fibrils reacted with the azide while maintaining the structure of the fibril. These click-functionalised amyloid fibrils have a variety of potential uses in materials and as scaffolds for bionanotechnology. DISCUSSION: Although previous studies have produced peptides that can both form amyloid fibrils and undergo "click"-type reactions, this is the first example of amyloid fibrils that can undergo such a reaction after they have been formed. Our approach has the advantage that self-assembly takes place before click functionalization rather than pre-functionalised building blocks self-assembling. Therefore, the molecules used to functionalise the fibril do not themselves have to be exposed to harsh, amyloid-forming conditions. This means that a wider range of proteins can be used as ligands in this process. For instance, the fibrils can be functionalised with a green fluorescent protein that retains its fluorescence after it is attached to the fibrils, whereas this protein loses its fluorescence if it is exposed to the conditions used for aggregation.


Subject(s)
Alkynes/chemistry , Amyloid/chemistry , Azides/chemistry , Click Chemistry/methods , Peptides/chemistry , Prealbumin/chemistry , Alkynes/chemical synthesis , Amino Acid Sequence , Amyloid/chemical synthesis , Azides/chemical synthesis , Green Fluorescent Proteins/chemical synthesis , Green Fluorescent Proteins/chemistry , Nanotechnology , Peptides/chemical synthesis , Prealbumin/chemical synthesis
7.
Anal Chem ; 89(18): 9976-9983, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28803470

ABSTRACT

Given the frequent use of DMSO in biochemical and biophysical assays, it is desirable to understand the influence of DMSO concentration on the dissociation or unfolding behavior of proteins. In this study, the effects of DMSO on the structure and interactions of avidin and Mycobacterium tuberculosis (Mtb) CYP142A1 were assessed through collision-induced dissociation (CID) and collision-induced unfolding (CIU) as monitored by nanoelectrospray ionization-ion mobility-mass spectrometry (nESI-IM-MS). DMSO concentrations higher than 4% (v/v) destabilize the avidin tetramer toward dissociation and unfolding, via both its effects on charge state distribution (CSD) as well as at the level of individual charge states. In contrast, DMSO both protects against heme loss and increases the stability of CYP142A1 toward unfolding even up to 40% DMSO. Tandem MS/MS experiments showed that DMSO could modify the dissociation pathway of CYP142A1, while CIU revealed the protective effect of the heme group on the structure of CYP142A1.


Subject(s)
Avidin/chemistry , Cytochrome P-450 Enzyme System/chemistry , Dimethyl Sulfoxide/pharmacology , Mycobacterium tuberculosis/enzymology , Cytochrome P-450 Enzyme System/metabolism , Dimethyl Sulfoxide/chemistry , Protein Conformation , Protein Unfolding , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
8.
Biomacromolecules ; 18(10): 3052-3059, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28792742

ABSTRACT

In nature, a wide range of functional materials is based on proteins. Increasing attention is also turning to the use of proteins as artificial biomaterials in the form of films, gels, particles, and fibrils that offer great potential for applications in areas ranging from molecular medicine to materials science. To date, however, most such applications have been limited to single component materials despite the fact that their natural analogues are composed of multiple types of proteins with a variety of functionalities that are coassembled in a highly organized manner on the micrometer scale, a process that is currently challenging to achieve in the laboratory. Here, we demonstrate the fabrication of multicomponent protein microcapsules where the different components are positioned in a controlled manner. We use molecular self-assembly to generate multicomponent structures on the nanometer scale and droplet microfluidics to bring together the different components on the micrometer scale. Using this approach, we synthesize a wide range of multiprotein microcapsules containing three well-characterized proteins: glucagon, insulin, and lysozyme. The localization of each protein component in multishell microcapsules has been detected by labeling protein molecules with different fluorophores, and the final three-dimensional microcapsule structure has been resolved by using confocal microscopy together with image analysis techniques. In addition, we show that these structures can be used to tailor the release of such functional proteins in a sequential manner. Moreover, our observations demonstrate that the protein release mechanism from multishell capsules is driven by the kinetic control of mass transport of the cargo and by the dissolution of the shells. The ability to generate artificial materials that incorporate a variety of different proteins with distinct functionalities increases the breadth of the potential applications of artificial protein-based materials and provides opportunities to design more refined functional protein delivery systems.


Subject(s)
Capsules/chemistry , Drug Liberation , Glucagon/chemistry , Insulin/chemistry , Muramidase/chemistry
9.
Angew Chem Int Ed Engl ; 56(26): 7488-7491, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28513917

ABSTRACT

Native nanoelectrospray ionization mass spectrometry is an underutilized technique for fragment screening. In this study, the first demonstration is provided of the use of native mass spectrometry for screening fragments against a protein-DNA interaction. EthR is a transcriptional repressor of EthA expression in Mycobacterium tuberculosis (Mtb) that reduces the efficacy of ethionamide, a second-line antitubercular drug used to combat multidrug-resistant Mtb strains. A small-scale fragment screening campaign was conducted against the EthR-DNA interaction using native mass spectrometry, and the results were compared with those from differential scanning fluorimetry, a commonly used primary screening technique. Hits were validated by surface plasmon resonance and X-ray crystallography. The screening campaign identified two new fragments that disrupt the EthR-DNA interaction in vitro (IC50 =460-610 µm) and bind to the hydrophobic channel of the EthR dimer.


Subject(s)
DNA/chemistry , Mass Spectrometry/methods , Mycobacterium tuberculosis/chemistry , Repressor Proteins/chemistry , Crystallography, X-Ray , Fluorometry/methods , Hydrophobic and Hydrophilic Interactions , Protein Conformation , Proteins/chemistry , Surface Plasmon Resonance
10.
Nucleic Acids Res ; 45(9): 5555-5563, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28334766

ABSTRACT

Human Timeless is involved in replication fork stabilization, S-phase checkpoint activation and establishment of sister chromatid cohesion. In the cell, Timeless forms a constitutive heterodimeric complex with Tipin. Here we present the 1.85 Å crystal structure of a large N-terminal segment of human Timeless, spanning amino acids 1-463, and we show that this region of human Timeless harbours a partial binding site for Tipin. Furthermore, we identify minimal regions of the two proteins that are required for the formation of a stable Timeless-Tipin complex and provide evidence that the Timeless-Tipin interaction is based on a composite binding interface comprising different domains of Timeless.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Biophysical Phenomena , Cross-Linking Reagents/metabolism , Crystallography, X-Ray , DNA-Binding Proteins , Humans , Mass Spectrometry , Models, Molecular , Protein Binding , Protein Domains , Protein Multimerization , Structural Homology, Protein
11.
Chem Commun (Camb) ; 53(25): 3527-3530, 2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28287239

ABSTRACT

EthR is a transcriptional repressor that increases Mycobacterium tuberculosis resistance to ethionamide. In this study, the EthR-DNA interaction has been investigated by native electrospray-ionization mass spectrometry for the first time. The results show that up to six subunits of EthR are able to bind to its operator.


Subject(s)
DNA, Bacterial/metabolism , Ethionamide/metabolism , Calorimetry , DNA, Bacterial/chemistry , Drug Resistance, Bacterial , Ethionamide/chemistry , Mycobacterium tuberculosis/genetics , Spectrometry, Mass, Electrospray Ionization , Thermodynamics
12.
J Am Chem Soc ; 139(8): 3202-3208, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28198190

ABSTRACT

A 1:1 binding stoichiometry of a host-guest complex need not consist of a single host and guest. Diarylviologens containing electron-donating substituents complexed with cucurbit[8]uril (CB[8]) in a 1:1 stoichiometry exhibit abnormally large binding enthalpies compared to typical enthalpy changes observed for 1:1 binary complexes. Here, several CB[8]-mediated host-guest complexes, which were previously reported as 1:1 binary complexes, are verified to be 2:2 quaternary complexes by a combination of isothermal titration calorimetry, 1H, NOESY, and ROESY NMR, and ion mobility mass spectrometry, clearly indicating a binding motif of two partially overlapping diarylviologens held in place with two CB[8] molecules. Formation of 2:2 quaternary complexes is favored by electron-donating substituents, while electron-withdrawing substituents typically result in 1:1 binary complexes. The stacking of two highly conjugated diarylviologens in one quaternary motif affords the complexes enhanced conductance when considered as a single-molecular conductor. Moreover, an additional conducting signal previously observed for this "supramolecular" conductor can be readily understood with our 2:2 complexation model, corresponding to a parallel conductance pathway. Therefore, a 2:2 quaternary complex model grants a greater understanding of such supramolecular complexes, enabling the design of engineered, hierarchical structures and functional materials.

13.
Mol Cell ; 63(3): 385-96, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27397685

ABSTRACT

Replisome assembly at eukaryotic replication forks connects the DNA helicase to DNA polymerases and many other factors. The helicase binds the leading-strand polymerase directly, but is connected to the Pol α lagging-strand polymerase by the trimeric adaptor Ctf4. Here, we identify new Ctf4 partners in addition to Pol α and helicase, all of which contain a "Ctf4-interacting-peptide" or CIP-box. Crystallographic analysis classifies CIP-boxes into two related groups that target different sites on Ctf4. Mutations in the CIP-box motifs of the Dna2 nuclease or the rDNA-associated protein Tof2 do not perturb DNA synthesis genome-wide, but instead lead to a dramatic shortening of chromosome 12 that contains the large array of rDNA repeats. Our data reveal unexpected complexity of Ctf4 function, as a hub that connects multiple accessory factors to the replisome. Most strikingly, Ctf4-dependent recruitment of CIP-box proteins couples other processes to DNA synthesis, including rDNA copy-number regulation.


Subject(s)
Chromosomes, Fungal/enzymology , DNA Helicases/metabolism , DNA, Fungal/biosynthesis , DNA, Ribosomal/biosynthesis , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , S Phase , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Chromosomes, Fungal/genetics , DNA Helicases/genetics , DNA Polymerase I/metabolism , DNA, Fungal/genetics , DNA, Ribosomal/genetics , DNA-Binding Proteins/genetics , Gene Dosage , Intracellular Signaling Peptides and Proteins/genetics , Models, Molecular , Multiprotein Complexes , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship
14.
Sci Rep ; 6: 26628, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225995

ABSTRACT

Mycobacterium tuberculosis (Mtb) causes the disease tuberculosis (TB). The virulent Mtb H37Rv strain encodes 20 cytochrome P450 (CYP) enzymes, many of which are implicated in Mtb survival and pathogenicity in the human host. Bioinformatics analysis revealed that CYP144A1 is retained exclusively within the Mycobacterium genus, particularly in species causing human and animal disease. Transcriptomic annotation revealed two possible CYP144A1 start codons, leading to expression of (i) a "full-length" 434 amino acid version (CYP144A1-FLV) and (ii) a "truncated" 404 amino acid version (CYP144A1-TRV). Computational analysis predicted that the extended N-terminal region of CYP144A1-FLV is largely unstructured. CYP144A1 FLV and TRV forms were purified in heme-bound states. Mass spectrometry confirmed production of intact, His6-tagged forms of CYP144A1-FLV and -TRV, with EPR demonstrating cysteine thiolate coordination of heme iron in both cases. Hydrodynamic analysis indicated that both CYP144A1 forms are monomeric. CYP144A1-TRV was crystallized and the first structure of a CYP144 family P450 protein determined. CYP144A1-TRV has an open structure primed for substrate binding, with a large active site cavity. Our data provide the first evidence that Mtb produces two different forms of CYP144A1 from alternative transcripts, with CYP144A1-TRV generated from a leaderless transcript lacking a 5'-untranslated region and Shine-Dalgarno ribosome binding site.


Subject(s)
Bacterial Proteins , Cytochrome P-450 Enzyme System , Mycobacterium tuberculosis , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Mass Spectrometry , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Protein Domains
15.
ACS Chem Biol ; 11(6): 1511-7, 2016 06 17.
Article in English | MEDLINE | ID: mdl-26999075

ABSTRACT

CK2 is an intrinsically active protein kinase that is crucial for cellular viability. However, conventional kinase regulatory mechanisms do not apply to CK2, and its mode of regulation remains elusive. Interestingly, CK2 is known to undergo reversible ionic-strength-dependent oligomerization. Furthermore, a regulatory mechanism based on autoinhibitory oligomerization has been postulated on the basis of the observation of circular trimeric oligomers and linear CK2 assemblies in various crystal structures. Here, we employ native mass spectrometry to monitor the assembly of oligomeric CK2 species in an ionic-strength-dependent manner. A subsequent combination of ion mobility spectrometry-mass spectrometry and hydrogen-deuterium exchange mass spectrometry techniques was used to analyze the conformation of CK2 oligomers. Our findings support ionic-strength-dependent CK2 oligomerization, demonstrate the transient nature of the α/ß interaction, and show that CK2 oligomerization proceeds via both the circular and linear assembly.


Subject(s)
Acetates/chemistry , Casein Kinase II/chemistry , Deuterium , Humans , Mass Spectrometry/methods , Osmolar Concentration , Protein Multimerization , Protein Structure, Quaternary
16.
J Mol Biol ; 427(12): 2166-78, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25861763

ABSTRACT

Huntington's disease is triggered by misfolding of fragments of mutant forms of the huntingtin protein (mHTT) with aberrant polyglutamine expansions. The C4 single-chain Fv antibody (scFv) binds to the first 17 residues of huntingtin [HTT(1-17)] and generates substantial protection against multiple phenotypic pathologies in situ and in vivo. We show in this paper that C4 scFv inhibits amyloid formation by exon1 fragments of huntingtin in vitro and elucidate the structural basis for this inhibition and protection by determining the crystal structure of the complex of C4 scFv and HTT(1-17). The peptide binds with residues 3-11 forming an amphipathic helix that makes contact with the antibody fragment in such a way that the hydrophobic face of this helix is shielded from the solvent. Residues 12-17 of the peptide are in an extended conformation and interact with the same region of another C4 scFv:HTT(1-17) complex in the asymmetric unit, resulting in a ß-sheet interface within a dimeric C4 scFv:HTT(1-17) complex. The nature of this scFv-peptide complex was further explored in solution by high-resolution NMR and physicochemical analysis of species in solution. The results provide insights into the manner in which C4 scFv inhibits the aggregation of HTT, and hence into its therapeutic potential, and suggests a structural basis for the initial interactions that underlie the formation of disease-associated amyloid fibrils by HTT.


Subject(s)
Amyloid/chemistry , Amyloid/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Amyloid/antagonists & inhibitors , Chemical Phenomena , Crystallography, X-Ray , Humans , Huntingtin Protein , Magnetic Resonance Spectroscopy , Models, Molecular , Nerve Tissue Proteins/antagonists & inhibitors , Protein Binding , Protein Multimerization , Protein Structure, Quaternary
17.
Chem Commun (Camb) ; 51(19): 3993-6, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25660271

ABSTRACT

We report the design and synthesis of an aquacarbonyl Ru(II) dication cis-[Ru(CO)2(H2O)4](2+) reagent for histidine (His)-selective metallation of interleukin (IL)-8 at site 33. The artificial, non-toxic interleukin (IL)-8-Ru(II)(CO)2 metalloprotein retained IL-8-dependent neutrophil chemotactic activity and was shown to spontaneously release CO in live cells.


Subject(s)
Biomimetic Materials/chemistry , Histidine/chemistry , Metalloproteins/chemistry , Ruthenium/chemistry , Chemotaxis , HeLa Cells , Humans , Interleukin-8/chemistry , Neutrophils/cytology
18.
Angew Chem Int Ed Engl ; 54(4): 1172-5, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25477186

ABSTRACT

We demonstrate that Ru(II)(CO)2-protein complexes, formed by the reaction of the hydrolytic decomposition products of [fac-RuCl(κ(2)-H2NCH2CO2)(CO)3] (CORM-3) with histidine residues exposed on the surface of proteins, spontaneously release CO in aqueous solution, cells, and mice. CO release was detected by mass spectrometry (MS) and confocal microscopy using a CO-responsive turn-on fluorescent probe. These findings support our hypothesis that plasma proteins act as CO carriers after in vivo administration of CORM-3. CO released from a synthetic bovine serum albumin (BSA)-Ru(II)(CO)2 complex leads to downregulation of the cytokines interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α in cancer cells. Finally, administration of BSA-Ru(II)(CO)2 in mice bearing a colon carcinoma tumor results in enhanced CO accumulation at the tumor. Our data suggest the use of Ru(II)(CO)2-protein complexes as viable alternatives for the safe and spatially controlled delivery of therapeutic CO in vivo.


Subject(s)
Organometallic Compounds/chemistry , Serum Albumin, Bovine/chemistry , Animals , Carbon Monoxide/analysis , Carbon Monoxide/metabolism , Cattle , Cell Line, Tumor , Female , HeLa Cells , Histidine/chemistry , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Mass Spectrometry , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Serum Albumin, Bovine/metabolism , Tissue Distribution , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/metabolism
19.
Nat Commun ; 5: 5165, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25387393

ABSTRACT

The transcription factor FOXM1 binds to sequence-specific motifs on DNA (C/TAAACA) through its DNA-binding domain (DBD) and activates proliferation- and differentiation-associated genes. Aberrant overexpression of FOXM1 is a key feature in oncogenesis and progression of many human cancers. Here--from a high-throughput screen applied to a library of 54,211 small molecules--we identify novel small molecule inhibitors of FOXM1 that block DNA binding. One of the identified compounds, FDI-6 (NCGC00099374), is characterized in depth and is shown to bind directly to FOXM1 protein, to displace FOXM1 from genomic targets in MCF-7 breast cancer cells, and induce concomitant transcriptional downregulation. Global transcript profiling of MCF-7 cells by RNA-seq shows that FDI-6 specifically downregulates FOXM1-activated genes with FOXM1 occupancy confirmed by ChIP-PCR. This small molecule-mediated effect is selective for FOXM1-controlled genes with no effect on genes regulated by homologous forkhead family factors.


Subject(s)
Forkhead Transcription Factors/antagonists & inhibitors , Pyridines/pharmacology , Thiophenes/pharmacology , Blotting, Western , Chromatin/metabolism , Down-Regulation/drug effects , Fluorescence Polarization , Forkhead Box Protein M1 , High-Throughput Screening Assays , Humans , MCF-7 Cells/drug effects , Transcription, Genetic/drug effects
20.
Nat Commun ; 5: 3902, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24852487

ABSTRACT

eIF2B facilitates and controls protein synthesis in eukaryotes by mediating guanine nucleotide exchange on its partner eIF2. We combined mass spectrometry (MS) with chemical cross-linking, surface accessibility measurements and homology modelling to define subunit stoichiometry and interactions within eIF2B and eIF2. Although it is generally accepted that eIF2B is a pentamer of five non-identical subunits (α-ε), here we show that eIF2B is a decamer. MS and cross-linking of eIF2B complexes allows us to propose a model for the subunit arrangements within eIF2B where the subunit assembly occurs through catalytic γ- and ε-subunits, with regulatory subunits arranged in asymmetric trimers associated with the core. Cross-links between eIF2 and eIF2B allow modelling of interactions that contribute to nucleotide exchange and its control by eIF2 phosphorylation. Finally, we identify that GTP binds to eIF2Bγ, prompting us to propose a multi-step mechanism for nucleotide exchange.


Subject(s)
Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2B/metabolism , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cross-Linking Reagents/pharmacology , Electrophoresis, Polyacrylamide Gel , Eukaryotic Initiation Factor-2/metabolism , Guanosine Triphosphate/metabolism , Hydrophobic and Hydrophilic Interactions , Mass Spectrometry , Models, Molecular , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Structure, Tertiary , Solvents , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...