Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 4(3): e1016, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38511507

ABSTRACT

Investigating the function of target proteins for functional prospection or therapeutic applications typically requires the production and purification of recombinant proteins. The fusion of these proteins with tag peptides and fluorescently derived proteins allows the monitoring of candidate proteins using SDS-PAGE coupled with western blotting and fluorescent microscopy, respectively. However, protein engineering poses a significant challenge for many researchers. In this protocol, we describe step-by-step the engineering of a recombinant protein with various tags: TAT-HA (trans-activator of transduction-hemagglutinin), 6×His and EGFP (enhanced green fluorescent protein) or mCherry. Fusion proteins are produced in E. coli BL21(DE3) cells and purified by immobilized metal affinity chromatography (IMAC) using a Ni-nitrilotriacetic acid (NTA) column. Then, tagged recombinant proteins are introduced into cultured animal cells by using the penetrating peptide TAT-HA. Here, we present a thorough protocol providing a detailed guide encompassing every critical step from plasmid DNA molecular assembly to protein expression and subsequent purification and outlines the conditions necessary for protein transduction technology into animal cells in a comprehensive manner. We believe that this protocol will be a valuable resource for researchers seeking an exhaustive, step-by-step guide for the successful production and purification of recombinant proteins and their entry by transduction within living cells. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: DNA cloning, molecular assembly strategies, and protein production Basic Protocol 2: Protein purification Basic Protocol 3: Protein transduction in mammalian cells.


Subject(s)
Escherichia coli , Peptides , Animals , Escherichia coli/genetics , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Peptides/genetics , Peptides/metabolism , Indicators and Reagents/metabolism , Gene Products, tat/metabolism , Coloring Agents/metabolism , DNA/metabolism , Mammals/genetics , Mammals/metabolism
2.
J Biol Chem ; 294(35): 13171-13185, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31315928

ABSTRACT

Implicated in numerous human diseases, intrinsically disordered proteins (IDPs) are dynamic ensembles of interconverting conformers that often contain many proline residues. Whether and how proline conformation regulates the functional aspects of IDPs remains an open question, however. Here, we studied the disordered domain 2 of nonstructural protein 5A (NS5A-D2) of hepatitis C virus (HCV). NS5A-D2 comprises a short structural motif (PW-turn) embedded in a proline-rich sequence, whose interaction with the human prolyl isomerase cyclophilin A (CypA) is essential for viral RNA replication. Using NMR, we show here that the PW-turn motif exists in a conformational equilibrium between folded and disordered states. We found that the fraction of conformers in the NS5A-D2 ensemble that adopt the structured motif is allosterically modulated both by the cis/trans isomerization of the surrounding prolines that are CypA substrates and by substitutions conferring resistance to cyclophilin inhibitor. Moreover, we noted that this fraction is directly correlated with HCV RNA replication efficiency. We conclude that CypA can fine-tune the dynamic ensemble of the disordered NS5A-D2, thereby regulating viral RNA replication efficiency.


Subject(s)
Cyclophilin A/metabolism , RNA, Viral/metabolism , Viral Nonstructural Proteins/metabolism , Allosteric Regulation , Cyclophilin A/genetics , Cyclophilin A/isolation & purification , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , RNA, Viral/chemistry , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/isolation & purification , Virus Replication
3.
ACS Chem Biol ; 14(6): 1363-1379, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31046227

ABSTRACT

Self-assembly of the microtubule-associated protein tau into neurotoxic oligomers, fibrils, and paired helical filaments, and cell-to-cell spreading of these pathological tau species are critical processes underlying the pathogenesis of Alzheimer's disease and other tauopathies. Modulating the self-assembly process and inhibiting formation and spreading of such toxic species are promising strategies for therapy development. A challenge in investigating tau self-assembly in vitro is that, unlike most amyloidogenic proteins, tau does not aggregate in the absence of posttranslational modifications (PTM), aggregation inducers, or preformed seeds. The most common induction method is addition of polyanions, such as heparin; yet, this artificial system may not represent adequately tau self-assembly in vivo, which is driven by aberrant phosphorylation and other PTMs, potentially leading to in vitro data that do not reflect the behavior of tau and its interaction with modulators in vivo. To tackle these challenges, methods for in vitro phosphorylation of tau to produce aggregation-competent forms recently have been introduced ( Despres et al. ( 2017 ) Proc. Natl. Acad. Sci. U.S.A. , 114 , 9080 - 9085 ). However, the oligomerization, seeding, and interaction with assembly modulators of the different forms of tau have not been studied to date. To address these knowledge gaps, we compared here side-by-side the self-assembly and seeding activity of heparin-induced tau with two forms of in vitro phosphorylated tau and tested how the molecular tweezer CLR01, a negatively charged compound, affected these processes. Tau was phosphorylated by incubation either with activated extracellular signal-regulated kinase 2 or with a whole rat brain extract. Seeding activity was measured using a fluorescence-resonance energy transfer-based biosensor-cell method. We also used solution-state NMR to investigate the binding sites of CLR01 on tau and how they were impacted by phosphorylation. Our systematic structure-activity relationship study demonstrates that heparin-induced tau behaves differently from in vitro phosphorylated tau. The aggregation rates of the different forms are distinct as is the intracellular localization of the induced aggregates, which resemble brain-derived tau strains suggesting that heparin-induced tau and in vitro phosphorylated tau have different conformations, properties, and activities. CLR01 inhibits aggregation and seeding of both heparin-induced and in vitro phosphorylated tau dose-dependently, although heparin induction interferes with the interaction between CLR01 and tau.


Subject(s)
Heparin/pharmacology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Humans , Phosphorylation , Rats , tau Proteins/antagonists & inhibitors
4.
Sci Rep ; 8(1): 13846, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218010

ABSTRACT

Tau is a Microtubule-associated protein that induces and stabilizes the formation of the Microtubule cytoskeleton and plays an important role in neurodegenerative diseases. The Microtubules binding region of Tau has been determined for a long time but where and how Tau binds to its partner still remain a topic of debate. We used Site Directed Spin Labeling combined with EPR spectroscopy to monitor Tau upon binding to either Taxol-stabilized MTs or to αß-tubulin when Tau is directly used as an inducer of MTs formation. Using maleimide-functionalized labels grafted on the two natural cysteine residues of Tau, we found in both cases that Tau remains highly flexible in these regions confirming the fuzziness of Tau:MTs complexes. More interestingly, using labels linked by a disulfide bridge, we evidenced for the first time thiol disulfide exchanges between αß-tubulin or MTs and Tau. Additionally, Tau fragments having the two natural cysteines or variants containing only one of them were used to determine the role of each cysteine individually. The difference observed in the label release kinetics between preformed MTs or Tau-induced MTs, associated to a comparison of structural data, led us to propose two putative binding sites of Tau on αß-tubulin.


Subject(s)
Disulfides/metabolism , Sulfhydryl Compounds/metabolism , Tubulin/chemistry , Tubulin/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Binding Sites , Microtubules/metabolism , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Quaternary
5.
J Am Chem Soc ; 140(26): 8138-8146, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29708745

ABSTRACT

In Alzheimer's disease, amyloid-ß (Aß) plaques and tau neurofibrillary tangles are the two pathological hallmarks. The co-occurrence and combined reciprocal pathological effects of Aß and tau protein aggregation have been observed in animal models of the disease. However, the molecular mechanism of their interaction remain unknown. Using a variety of biophysical measurements, we here show that the native full-length tau protein solubilizes the Aß40 peptide and prevents its fibrillation. The tau protein delays the amyloid fibrillation of the Aß40 peptide at substoichiometric ratios, showing different binding affinities toward the different stages of the aggregated Aß40 peptides. The Aß monomer structure remains random coil in the presence of tau, as observed by nuclear magnetic resonance (NMR), circular dichroism (CD) spectroscopy and photoinduced cross-linking methods. We propose a potential interaction mechanism for the influence of tau on Aß fibrillation.


Subject(s)
Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/chemistry , Circular Dichroism , Humans , Microscopy, Atomic Force , Nuclear Magnetic Resonance, Biomolecular , Particle Size , Surface Properties , tau Proteins/chemistry
6.
Proc Natl Acad Sci U S A ; 114(34): 9080-9085, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28784767

ABSTRACT

Determining the functional relationship between Tau phosphorylation and aggregation has proven a challenge owing to the multiple potential phosphorylation sites and their clustering in the Tau sequence. We use here in vitro kinase assays combined with NMR spectroscopy as an analytical tool to generate well-characterized phosphorylated Tau samples and show that the combined phosphorylation at the Ser202/Thr205/Ser208 sites, together with absence of phosphorylation at the Ser262 site, yields a Tau sample that readily forms fibers, as observed by thioflavin T fluorescence and electron microscopy. On the basis of conformational analysis of synthetic phosphorylated peptides, we show that aggregation of the samples correlates with destabilization of the turn-like structure defined by phosphorylation of Ser202/Thr205.


Subject(s)
Protein Aggregation, Pathological , Serine/metabolism , Threonine/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Humans , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Phosphorylation , Protein Domains , Rats, Sprague-Dawley , Serine/chemistry , Serine/genetics , Threonine/chemistry , Threonine/genetics , tau Proteins/chemistry , tau Proteins/genetics
7.
Biophys J ; 112(5): 921-932, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28297651

ABSTRACT

Tau aggregates into paired helical filaments within neurons, a pathological hallmark of Alzheimer's disease. Heparin promotes tau aggregation and recently has been shown to be involved in the cellular uptake of tau aggregates. Although the tau-heparin interaction has been extensively studied, little is known about the glycan determinants of this interaction. Here, we used surface plasmon resonance (SPR) and NMR spectroscopy to characterize the interaction between two tau fragments, K18 and K19, and several polysaccharides, including heparin, heparin oligosaccharides, chemically modified heparin, and related glycans. Using a heparin-immobilized chip, SPR revealed that tau K18 and K19 bind heparin with a KD of 0.2 and 70 µM, respectively. In SPR competition experiments, N-desulfation and 2-O-desulfation had no effect on heparin binding to K18, whereas 6-O-desulfation severely reduced binding, suggesting a critical role for 6-O-sulfation in the tau-heparin interaction. The tau-heparin interaction became stronger with longer-chain heparin oligosaccharides. As expected for an electrostatics-driven interaction, a moderate amount of salt (0.3 M NaCl) abolished binding. NMR showed the largest chemical-shift perturbation (CSP) in R2 in tau K18, which was absent in K19, revealing differential binding sites in K18 and K19 to heparin. Dermatan sulfate binding produced minimal CSP, whereas dermatan disulfate, with the additional 6-O-sulfo group, induced much larger CSP. 2-O-desulfated heparin induced much larger CSP in K18 than 6-O-desulfated heparin. Our data demonstrate a crucial role for the 6-O-sulfo group in the tau-heparin interaction, which to our knowledge has not been reported before.


Subject(s)
Heparin/chemistry , Heparin/metabolism , tau Proteins/metabolism , Dose-Response Relationship, Drug , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/drug effects , Protein Conformation , Sodium Chloride/pharmacology , Surface Plasmon Resonance , tau Proteins/chemistry
8.
Biomolecules ; 6(2)2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27338491

ABSTRACT

In this review, we focus on what we have learned from Nuclear Magnetic Resonance (NMR) studies on the neuronal microtubule-associated protein Tau. We consider both the mechanistic details of Tau: the tubulin relationship and its aggregation process. Phosphorylation of Tau is intimately linked to both aspects. NMR spectroscopy has depicted accurate phosphorylation patterns by different kinases, and its non-destructive character has allowed functional assays with the same samples. Finally, we will discuss other post-translational modifications of Tau and its interaction with other cellular factors in relationship to its (dys)function.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , tau Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Protein Processing, Post-Translational , Tubulin/chemistry , Tubulin/metabolism , tau Proteins/metabolism
9.
Angew Chem Int Ed Engl ; 55(26): 7418-22, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27159340

ABSTRACT

Under physiological conditions, studies of intrinsically disordered proteins (IDPs) by conventional NMR methods based on proton detection are severely limited by fast amide-proton exchange with water. (13) C detection has been proposed as a solution to the exchange problem, but is hampered by low sensitivity. We propose a new pulse sequence combining proton-nitrogen cross-polarization and carbonyl detection to record high-resolution, high-sensitivity NMR spectra of IDPs under physiological conditions. To demonstrate the efficacy of this approach, we recorded a high-quality N-CO correlation spectrum of α-synuclein in bacterial cells at 37 °C.


Subject(s)
Intrinsically Disordered Proteins/chemistry , alpha-Synuclein/chemistry , Bacteria , Magnetic Resonance Spectroscopy , Nitrogen/chemistry , Protein Carbonylation
10.
J Vis Exp ; (118)2016 12 27.
Article in English | MEDLINE | ID: mdl-28060278

ABSTRACT

Aggregates of the neuronal Tau protein are found inside neurons of Alzheimer's disease patients. Development of the disease is accompanied by increased, abnormal phosphorylation of Tau. In the course of the molecular investigation of Tau functions and dysfunctions in the disease, nuclear magnetic resonance (NMR) spectroscopy is used to identify the multiple phosphorylations of Tau. We present here detailed protocols of recombinant production of Tau in bacteria, with isotopic enrichment for NMR studies. Purification steps that take advantage of Tau's heat stability and high isoelectric point are described. The protocol for in vitro phosphorylation of Tau by recombinant activated ERK2 allows for generating multiple phosphorylations. The protein sample is ready for data acquisition at the issue of these steps. The parameter setup to start recording on the spectrometer is considered next. Finally, the strategy to identify phosphorylation sites of modified Tau, based on NMR data, is explained. The benefit of this methodology compared to other techniques used to identify phosphorylation sites, such as immuno-detection or mass spectrometry (MS), is discussed.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Magnetic Resonance Spectroscopy , Phosphorylation , tau Proteins/chemistry , Alzheimer Disease , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry
11.
J Mol Biol ; 428(1): 79-91, 2016 Jan 16.
Article in English | MEDLINE | ID: mdl-26655856

ABSTRACT

The conformational state of distinct prolines can determine the folding of a protein but equally other biological processes when coupled to a conformation-sensitive secondary reaction. For the neuronal tau protein, the importance of proline conformation is underscored by its interaction with different prolyl cis/trans isomerases. The proline conformation would gain even further importance after phosphorylation of the preceding residue by various proline-directed kinases. A number of molecular diseases including Alzheimer's disease and traumatic brain injury were thereby recently qualified as "cistauosis", as they would imply a cis conformation for the pThr231-Pro232 prolyl bond. We here investigate by NMR spectroscopy the conformation of all prolines in a functional Tau fragment, Tau[208-324]. Although we can detect and identify some minor conformers in the cis form, we show that all prolines are for over 90% in the trans conformation. Phosphorylation by CDK2/CycA3, which notably leads to complete modification of the Thr231 residue, does not change this conclusion. Our data hence disagree with the notion that specific prolyl bonds in tau would adopt preferentially the cis conformation.


Subject(s)
Proline/chemistry , Proline/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Humans , Magnetic Resonance Spectroscopy , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational
12.
J Biol Chem ; 290(31): 19104-20, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26085105

ABSTRACT

Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) and its interaction with the human chaperone cyclophilin A are both targets for highly potent and promising antiviral drugs that are in the late stages of clinical development. Despite its high interest in regards to the development of drugs to counteract the worldwide HCV burden, NS5A is still an enigmatic multifunctional protein poorly characterized at the molecular level. NS5A is required for HCV RNA replication and is involved in viral particle formation and regulation of host pathways. Thus far, no enzymatic activity or precise molecular function has been ascribed to NS5A that is composed of a highly structured domain 1 (D1), as well as two intrinsically disordered domains 2 (D2) and 3 (D3), representing half of the protein. Here, we identify a short structural motif in the disordered NS5A-D2 and report its NMR structure. We show that this structural motif, a minimal Pro(314)-Trp(316) turn, is essential for HCV RNA replication, and its disruption alters the subcellular distribution of NS5A. We demonstrate that this Pro-Trp turn is required for proper interaction with the host cyclophilin A and influences its peptidyl-prolyl cis/trans isomerase activity on residue Pro(314) of NS5A-D2. This work provides a molecular basis for further understanding of the function of the intrinsically disordered domain 2 of HCV NS5A protein. In addition, our work highlights how very small structural motifs present in intrinsically disordered proteins can exert a specific function.


Subject(s)
Hepacivirus/enzymology , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/chemistry , Amino Acid Motifs , Cyclophilin A/chemistry , Humans , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Proline/chemistry , RNA, Viral/genetics , Tryptophan/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication
13.
PLoS One ; 10(6): e0129965, 2015.
Article in English | MEDLINE | ID: mdl-26090800

ABSTRACT

Delta-lactoferrin is a transcription factor, the expression of which is downregulated or silenced in case of breast cancer. It possesses antitumoral activities and when it is re-introduced in mammary epithelial cancer cell lines, provokes antiproliferative effects. It is posttranslationally modified and our earlier investigations showed that the O-GlcNAcylation/phosphorylation interplay plays a major role in the regulation of both its stability and transcriptional activity. Here, we report the covalent modification of delta-lactoferrin with the small ubiquitin-like modifier SUMO-1. Mutational and reporter gene analyses identified five different lysine residues at K13, K308, K361, K379 and K391 as SUMO acceptor sites. The SUMOylation deficient M5S mutant displayed enhanced transactivation capacity on a delta-lactoferrin responsive promoter, suggesting that SUMO-1 negatively regulates the transactivation function of delta-lactoferrin. K13, K308 and K379 are the main SUMO sites and among them, K308, which is located in a SUMOylation consensus motif of the NDSM-like type, is a key SUMO site involved in repression of delta-lactoferrin transcriptional activity. K13 and K379 are both targeted by other posttranslational modifications. We demonstrated that K13 is the main acetylation site and that favoring acetylation at K13 reduced SUMOylation and increased delta-lactoferrin transcriptional activity. K379, which is either ubiquitinated or SUMOylated, is a pivotal site for the control of delta-lactoferrin stability. We showed that SUMOylation competes with ubiquitination and protects delta-lactoferrin from degradation by positively regulating its stability. Collectively, our results indicate that multi-SUMOylation occurs on delta-lactoferrin to repress its transcriptional activity. Reciprocal occupancy of K13 by either SUMO-1 or an acetyl group may contribute to the establishment of finely regulated mechanisms to control delta-lactoferrin transcriptional activity. Moreover, competition between SUMOylation and ubiquitination at K379 coordinately regulates the stability of delta-lactoferrin toward proteolysis. Therefore SUMOylation of delta-lactoferrin is a novel mechanism controlling both its activity and stability.


Subject(s)
Lactoferrin/genetics , Lactoferrin/metabolism , Sumoylation , Transcriptional Activation , Acetylation , Amino Acid Motifs , Cell Line , Gene Expression , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Reporter , Humans , Lactoferrin/chemistry , Mutation , Protein Interaction Domains and Motifs , Protein Stability , Transcription Factors , Ubiquitination
14.
FASEB J ; 29(8): 3171-81, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25888602

ABSTRACT

Tauopathies, including Alzheimer's disease (AD), are neurodegenerative diseases associated with the pathologic aggregation of human brain Tau protein. Neuronal Tau is involved in microtubule (MT) formation and stabilization. We showed previously that the immunophilin FK506-binding protein of MW ∼52 kDa (FKBP52) interferes with this function of full-length Tau and provokes aggregation of a disease-related mutant of Tau. To dissect the molecular interaction between recombinant human FKBP52 and Tau, here, we study the effect of FKBP52 on a functional Tau fragment (Tau-F4, Ser(208)-Ser(324)) containing part of the proline- rich region and MT-binding repeats. Therefore, we perform MT assembly and light-scattering assays, blue native PAGE analysis, electron microscopy, and Tau seeding experiments in SH-SY5Y human neuroblastoma cells. We show that FKBP52 (6 µM) prevents MT formation generated by Tau-F4 (5 µM) and induces Tau-F4 oligomerization and aggregation. Electron microscopy analyses show granular oligomers and filaments of Tau-F4 after short-time FKBP52 incubation. We demonstrate that the terminal parts of Tau interfere with the effects of FKBP52. Finally, we find that FKBP52-induced Tau-F4 oligomers cannot only generate in vitro, direct conformational changes in full-length Tau and that their uptake into neuronal cells can equally lead to aggregation of wild-type endogenous Tau. This suggests a potential prion-like property of these particular Tau-F4 aggregates. Collectively, our results strengthen the hypothesis of FKBP52 involvement in the Tau pathogenicity process.


Subject(s)
Prions/metabolism , Protein Binding/physiology , Tacrolimus Binding Proteins/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Humans , Male , Microtubules/metabolism , Rats , Rats, Sprague-Dawley , Tauopathies/metabolism
15.
J Biol Chem ; 290(7): 4059-74, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25540200

ABSTRACT

A link between Tau phosphorylation and aggregation has been shown in different models for Alzheimer disease, including yeast. We used human Tau purified from yeast models to generate new monoclonal antibodies, of which three were further characterized. The first antibody, ADx201, binds the Tau proline-rich region independently of the phosphorylation status, whereas the second, ADx215, detects an epitope formed by the Tau N terminus when Tau is not phosphorylated at Tyr(18). For the third antibody, ADx210, the binding site could not be determined because its epitope is probably conformational. All three antibodies stained tangle-like structures in different brain sections of THY-Tau22 transgenic mice and Alzheimer patients, and ADx201 and ADx210 also detected neuritic plaques in the cortex of the patient brains. In hippocampal homogenates from THY-Tau22 mice and cortex homogenates obtained from Alzheimer patients, ADx215 consistently stained specific low order Tau oligomers in diseased brain, which in size correspond to Tau dimers. ADx201 and ADx210 additionally reacted to higher order Tau oligomers and presumed prefibrillar structures in the patient samples. Our data further suggest that formation of the low order Tau oligomers marks an early disease stage that is initiated by Tau phosphorylation at N-terminal sites. Formation of higher order oligomers appears to require additional phosphorylation in the C terminus of Tau. When used to assess Tau levels in human cerebrospinal fluid, the antibodies permitted us to discriminate patients with Alzheimer disease or other dementia like vascular dementia, indicative that these antibodies hold promising diagnostic potential.


Subject(s)
Alzheimer Disease/diagnosis , Antibodies, Monoclonal , Brain/pathology , Hippocampus/pathology , tau Proteins/chemistry , tau Proteins/immunology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/immunology , Animals , Biotinylation , Blotting, Western , Brain/immunology , Brain/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Hippocampus/immunology , Hippocampus/metabolism , Humans , Immunization , Immunoenzyme Techniques , Immunoprecipitation , Magnetic Resonance Spectroscopy , Membrane Microdomains , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neurofibrillary Tangles , Peptide Fragments/metabolism , Phosphorylation , Plaque, Amyloid , Saccharomyces cerevisiae , tau Proteins/cerebrospinal fluid
16.
J Magn Reson ; 249: 32-37, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25462944

ABSTRACT

We present an equilibrium H/D exchange experiment to measure the exchange rates of labile amide protons in intrinsically unfolded proteins. By measuring the contribution of the H/D exchange to the apparent T1 relaxation rates in solvents of different D2O content, we can easily derive the rates of exchange for rapidly exchanging amide protons. The method does not require double isotope labelling, is sensitive, and requires limited fitting of the data. We demonstrate it on a functional fragment of Tau, and provide evidence for the hydrogen bond formation of the phosphate moiety of Ser214 with its own amide proton in the same fragment phosphorylated by the PKA kinase.

17.
J Am Chem Soc ; 136(36): 12615-23, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25162583

ABSTRACT

Determining the molecular mechanism of the neuronal Tau protein in the tubulin heterodimer assembly has been a challenge owing to the dynamic character of the complex and the large size of microtubules. We use here defined constructs comprising one or two tubulin heterodimers to characterize their association with a functional fragment of Tau, named TauF4. TauF4 binds with high affinities to the tubulin heterodimer complexes, but NMR spectroscopy shows that it remains highly dynamic, partly because of the interaction with the acidic C-terminal tails of the tubulin monomers. When bound to a single tubulin heterodimer, TauF4 is characterized by an overhanging peptide corresponding to the first of the four microtubule binding repeats of Tau. This peptide becomes immobilized in the complex with two longitudinally associated tubulin heterodimers. The longitudinal associations are favored by the fragment and contribute to Tau's functional role in microtubule assembly.


Subject(s)
Microtubules/metabolism , Nuclear Magnetic Resonance, Biomolecular , tau Proteins/metabolism , Microtubules/chemistry , Models, Molecular , Tubulin/chemistry , Tubulin/metabolism , tau Proteins/chemistry
18.
PLoS One ; 9(8): e104563, 2014.
Article in English | MEDLINE | ID: mdl-25116916

ABSTRACT

BACKGROUND: Lactoferrins exhibit antitumoral activities either as a secretory lactoferrin or an intracellular delta-lactoferrin isoform. These activities involve processes such as regulation of the cell cycle and apoptosis. While lactoferrin has been shown to exert its function by activating different transduction pathways, delta-lactoferrin has been proven to act as a transcription factor. Like many tumor suppressors, these two proteins are under-expressed in several types of cancer, particularly in breast cancer. METHODOLOGY/PRINCIPAL FINDINGS: In order to compare the differential effects of the re-introduction of lactoferrin isoforms in breast cancer cells we chose the cancerous mammary gland MDA-MB-231 cell line as a model. We produced a cell line stably expressing delta-lactoferrin. We also treated these cells with fresh purified human breast lactoferrin. We performed two quantitative proteomic studies in parallel using SILAC coupled to mass spectrometry in order to compare the effects of different doses of the two lactoferrin isoforms. The proteome of untreated, delta-lactoferrin expressing and human lactoferrin treated MDA-MB-231 cells were compared. Overall, around 5300 proteins were identified and quantified using the in-house developed MFPaQ software. Among these, expression was increased by 1.5-fold or more for around 300 proteins in delta-lactoferrin expressing cells and 190 proteins in lactoferrin treated cells. At the same time, about 200 and 40 proteins were found to be downregulated (0-0.7-fold) in response to delta-lactoferrin and lactoferrin, respectively. CONCLUSIONS/SIGNIFICANCE: Re-introduction of delta-lactoferrin and lactoferrin expression in MDA-MB-231 mainly leads to modifications of protein profiles involved in processes such as proliferation, apoptosis, oxidative stress, the ubiquitin pathway, translation and mRNA quality control. Moreover, this study identified new target genes of delta-lactoferrin transcriptional activity such as SelH, GTF2F2 and UBE2E1.


Subject(s)
Lactoferrin/metabolism , Proteomics , Alternative Splicing , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Isotope Labeling , Lactoferrin/genetics , Lactoferrin/pharmacology , Neoplasm Metastasis , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Proteome , Proteomics/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reproducibility of Results , Response Elements , Selenoproteins/genetics , Transcription Factors/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
19.
Biochemistry ; 53(18): 3020-32, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24708343

ABSTRACT

Lysine acetylation of the neuronal Tau protein was described as a novel mechanism of posttranslational regulation of Tau functions with important outcomes in microtubule binding and aggregation processes related to Alzheimer's disease. Here, we unravel at a per-residue resolution the acetylation pattern of full-length Tau by the Creb-binding protein (CBP) acetyltransferase using high-resolution nuclear magnetic resonance spectroscopy. Our study gives a quantitative overview of CBP-mediated acetylation and examines the catalytic proficiency because the nonenzymatic reaction with acetyl-coenzyme A occurs in vitro. Furthermore, we have investigated with this characterized acetylated Tau the effect of acetylation on Tau fibrillization in a heparin-induced aggregation assay and on heparin binding.


Subject(s)
p300-CBP Transcription Factors/metabolism , tau Proteins/metabolism , Acetylation , Cysteine/chemistry , Heparin/pharmacology , Humans , Kinetics , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Protein Processing, Post-Translational , tau Proteins/chemistry , tau Proteins/drug effects
20.
Biochem Biophys Res Commun ; 445(2): 299-303, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24502945

ABSTRACT

We study the aggregation of a fragment of the neuronal protein Tau that contains part of the proline rich domain and of the microtubule binding repeats. When incubated at 37 °C with heparin, the fragment readily forms fibers as witnessed by Thioflavin T fluorescence. Electron microscopy and NMR spectroscopy show bundled ribbon like structures with most residues rigidly incorporated in the fibril. Without its cysteines, this fragment still forms fibers of a similar morphology, but with lesser Thioflavin T binding sites and more mobility for the C-terminal residues.


Subject(s)
Cysteine/chemistry , tau Proteins/chemistry , tau Proteins/ultrastructure , Cysteine/metabolism , Heparin/metabolism , Humans , Magnetic Resonance Spectroscopy , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...