Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(34): e2204618119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969745

ABSTRACT

Occludin is a tetramembrane-spanning tight junction protein. The long C-terminal cytoplasmic domain, which represents nearly half of occludin sequence, includes a distal bundle of three α-helices that mediates interactions with other tight junction components. A short unstructured region just proximal to the α-helical bundle is a phosphorylation hotspot within which S408 phosphorylation acts as molecular switch that modifies tight junction protein interactions and barrier function. Here, we used NMR to define the effects of S408 phosphorylation on intramolecular interactions between the unstructured region and the α-helical bundle. S408 pseudophosphorylation affected conformation at hinge sites between the three α-helices. Further studies using paramagnetic relaxation enhancement and microscale thermophoresis indicated that the unstructured region interacts with the α-helical bundle. These interactions between the unstructured domain are enhanced by S408 phosphorylation and allow the unstructured region to obstruct the binding site, thereby reducing affinity of the occludin tail for zonula occludens-1 (ZO-1). Conversely, S408 dephosphorylation attenuates intramolecular interactions, exposes the binding site, and increases the affinity of occludin binding to ZO-1. Consistent with an increase in binding to ZO-1, intravital imaging and fluorescence recovery after photobleaching (FRAP) analyses of transgenic mice demonstrated increased tight junction anchoring of enhanced green fluorescent protein (EGFP)-tagged nonphosphorylatable occludin relative to wild-type EGFP-occludin. Overall, these data define the mechanisms by which S408 phosphorylation modifies occludin tail conformation to regulate tight junction protein interactions and paracellular permeability.


Subject(s)
Phosphoproteins , Serine , Animals , Mice , Occludin/genetics , Occludin/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Conformation, alpha-Helical , Serine/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
2.
J Am Chem Soc ; 143(30): 11462-11472, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34308630

ABSTRACT

Amyloid-ß (Aß) fibrils in neuritic plaques are a hallmark of Alzheimer's disease (AD). Since the 42-residue Aß (Aß42) fibril is the most pathogenic among different Aß species, its structural characterization is crucial to our understanding of AD. While several polymorphs have been reported for Aß40, previous studies of Aß42 fibrils prepared at neutral pH detected essentially only one structure, with an S-shaped ß-sheet arrangement (e.g., Xiao et al. Nat. Struct. Mol. Biol. 2015, 22, 499). Herein, we demonstrate the feasibility of characterizing the structure of trace amounts of brain-derived and synthetic amyloid fibrils by sensitivity-enhanced 1H-detected solid-state NMR (SSNMR) under ultrafast magic angle spinning. By taking advantage of the high sensitivity of this technique, we first demonstrate its applicability for the high-throughput screening of trace amounts of selectively 13C- and 15N-labeled Aß42 fibril prepared with ∼0.01% patient-derived amyloid (ca. 4 pmol) as a seed. The comparison of 2D 13C/1H SSNMR data revealed marked structural differences between AD-derived Aß42 (∼40 nmol or ∼200 µg) and synthetic fibrils in less than 10 min, confirming the feasibility of assessing the fibril structure from ∼1 pmol of brain amyloid seed in ∼2.5 h. We also present the first structural characterization of synthetic fully protonated Aß42 fibril by 1H-detected 3D and 4D SSNMR. With procedures assisted by automated assignments, main-chain resonance assignments were completed for trace amounts (∼42 nmol) of a fully protonated amyloid fibril in the 1H-detection approach. The results suggest that this Aß42 fibril exhibits a novel fold or polymorph structure.


Subject(s)
Amyloid beta-Peptides/chemistry , Nuclear Magnetic Resonance, Biomolecular , Amyloid beta-Peptides/metabolism , Brain/metabolism , Humans , Protein Conformation , Proteins
3.
Biochemistry ; 60(36): 2691-2703, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34029056

ABSTRACT

Using atomic force microscopy (AFM) and nuclear magnetic resonance (NMR), we describe small Aß40 oligomers, termed nanodroplet oligomers (NanDOs), which form rapidly and at Aß40 concentrations too low for fibril formation. NanDOs were observed in putatively monomeric solutions of Aß40 (e.g., by size exclusion chromatography). Video-rate scanning AFM shows rapid fusion and dissolution of small oligomer-sized particles, of which the median size increases with peptide concentration. In NMR (13C HSQC), a small number of chemical shifts changed with a change in peptide concentration. Paramagnetic relaxation enhancement NMR experiments also support the formation of NanDOs and suggest prominent interactions in hydrophobic domains of Aß40. Addition of Zn2+ to Aß40 solutions caused flocculation of NanDO-containing solutions, and selective loss of signal intensity in NMR spectra from residues in the N-terminal domain of Aß40. NanDOs may represent the earliest aggregated form of Aß40 in the aggregation pathway and are akin to premicelles in solutions of amphiphilies.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Magnetic Resonance Spectroscopy/methods , Microscopy, Atomic Force/methods , Nanoparticles/chemistry , Protein Aggregates/physiology , Alzheimer Disease/metabolism , Electron Spin Resonance Spectroscopy/methods , Humans
4.
Sci Rep ; 11(1): 247, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420184

ABSTRACT

Alzheimer's disease is characterized by neuritic plaques, the main protein components of which are ß-amyloid (Aß) peptides deposited as ß-sheet-rich amyloid fibrils. Cerebral Amyloid Angiopathy (CAA) consists of cerebrovascular deposits of Aß peptides; it usually accompanies Alzheimer's disease, though it sometimes occurs in the absence of neuritic plaques, as AD also occurs without accompanying CAA. Although neuritic plaques and vascular deposits have similar protein compositions, one of the characteristic features of amyloids is polymorphism, i.e., the ability of a single pure peptide to adopt multiple conformations in fibrils, depending on fibrillization conditions. For this reason, we asked whether the Aß fibrils in neuritic plaques differed structurally from those in cerebral blood vessels. To address this question, we used seeding techniques, starting with amyloid-enriched material from either brain parenchyma or cerebral blood vessels (using meninges as the source). These amyloid-enriched preparations were then added to fresh, disaggregated solutions of Aß to make replicate fibrils, as described elsewhere. Such fibrils were then studied by solid-state NMR, fiber X-ray diffraction, and other biophysical techniques. We observed chemical shift differences between parenchymal vs. vascular-seeded replicate fibrils in select sites (in particular, Ala2, Phe4, Val12, and Gln15 side chains) in two-dimensional 13C-13C correlation solid-state NMR spectra, strongly indicating structural differences at these sites. X-ray diffraction studies also indicated that vascular-seeded fibrils displayed greater order than parenchyma-seeded fibrils in the "side-chain dimension" (~ 10 Å reflection), though the "hydrogen-bond dimensions" (~ 5 Å reflection) were alike. These results indicate that the different nucleation conditions at two sites in the brain, parenchyma and blood vessels, affect the fibril products that get formed at each site, possibly leading to distinct pathophysiological outcomes.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Brain/blood supply , Brain/metabolism , Brain/cytology , Humans , Protein Aggregates , Protein Conformation, beta-Strand
5.
Epigenetics ; 16(12): 1317-1334, 2021 12.
Article in English | MEDLINE | ID: mdl-33356812

ABSTRACT

Because ADAM17 promotes colonic tumorigenesis, we investigated potential miRNAs regulating ADAM17; and examined effects of diet and tumorigenesis on these miRNAs. We also examined pre-miRNA processing and tumour suppressor roles of several of these miRNAs in experimental colon cancer. Using TargetScan, miR-145, miR-148a, and miR-152 were predicted to regulate ADAM17. miR-143 was also investigated as miR-143 and miR-145 are co-transcribed and associated with decreased tumour growth. HCT116 colon cancer cells (CCC) were co-transfected with predicted ADAM17-regulating miRNAs and luciferase reporters controlled by ADAM17-3'UTR. Separately, pre-miR-143 processing by colonic cells was measured. miRNAs were quantified by RT-PCR. Tumours were induced with AOM/DSS in WT and transgenic mice (Tg) expressing pre-miR-143/miR-145 under villin promoter. HCT116 transfection with miR-145, -148a or -152, but not scrambled miRNA inhibited ADAM17 expression and luciferase activity. The latter was suppressed by mutations in ADAM17-3'UTR. Lysates from colonocytes, but not CCC, processed pre-miR-143 and mixing experiments suggested CCC lacked a competency factor. Colonic miR-143, miR-145, miR-148a, and miR-152 were downregulated in tumours and more moderately by feeding mice a Western diet. Tg mice were resistant to DSS colitis and had significantly lower cancer incidence and tumour multiplicity. Tg expression blocked up-regulation of putative targets of miR-143 and miR-145, including ADAM17, K-Ras, XPO5, and SET. miR-145, miR-148a, and miR-152 directly suppress colonocyte ADAM17 and are down-regulated in colon cancer. This is the first direct demonstration of tumour suppressor roles for miR-143 and miR-145 in an in vivo model of colonic tumorigenesis.


Subject(s)
Colitis , Colonic Neoplasms , MicroRNAs , Animals , Colonic Neoplasms/genetics , DNA Methylation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Inflammation , Karyopherins , Mice , MicroRNAs/metabolism , Up-Regulation
6.
Protein Sci ; 29(2): 527-541, 2020 02.
Article in English | MEDLINE | ID: mdl-31710741

ABSTRACT

The mechanism by which a disordered peptide nucleates and forms amyloid is incompletely understood. A central domain of ß-amyloid (Aß21-30) has been proposed to have intrinsic structural propensities that guide the limited formation of structure in the process of fibrillization. In order to test this hypothesis, we examine several internal fragments of Aß, and variants of these either cyclized or with an N-terminal Cys. While Aß21-30 and variants were always monomeric and unstructured (circular dichroism (CD) and nuclear magnetic resonance spectroscopy (NMRS)), we found that the addition of flanking hydrophobic residues in Aß16-34 led to formation of typical amyloid fibrils. NMR showed no long-range nuclear overhauser effect (nOes) in Aß21-30, Aß16-34, or their variants, however. Serial 1 H-15 N-heteronuclear single quantum coherence spectroscopy, 1 H-1 H nuclear overhauser effect spectroscopy, and 1 H-1 H total correlational spectroscopy spectra were used to follow aggregation of Aß16-34 and Cys-Aß16-34 at a site-specific level. The addition of an N-terminal Cys residue (in Cys-Aß16-34) increased the rate of fibrillization which was attributable to disulfide bond formation. We propose a scheme comparing the aggregation pathways for Aß16-34 and Cys-Aß16-34, according to which Cys-Aß16-34 dimerizes, which accelerates fibril formation. In this context, cysteine residues form a focal point that guides fibrillization, a role which, in native peptides, can be assumed by heterogeneous nucleators of aggregation.


Subject(s)
Amyloid beta-Peptides/chemistry , Disulfides/chemistry , Amyloid beta-Peptides/chemical synthesis , Hydrophobic and Hydrophilic Interactions
8.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31672936

ABSTRACT

Human cancer cells were eradicated by adoptive transfer of T cells transduced with a chimeric antigen receptor (CAR) made from an antibody (237Ab) that is highly specific for the murine Tn-glycosylated podoplanin (Tn-PDPN). The objectives were to determine the specificity of these CAR-transduced T (CART) cells and the mechanism for the absence of relapse. We show that although the 237Ab bound only to cell lines expressing murine Tn-PDPN, the 237Ab-derived 237CART cells lysed multiple different human and murine cancers not predicted by the 237Ab binding. Nevertheless, the 237CART cell reactivities remained cancer specific because all recognitions were dependent on the Tn glycosylation that resulted from COSMC mutations that were not present in normal tissues. While Tn was required for the recognition by 237CART, Tn alone was not sufficient for 237CART cell activation. Activation of 237CART cells required peptide backbone recognition but tolerated substitutions of up to 5 of the 7 amino acid residues in the motif recognized by 237Ab. Together, these findings demonstrate what we believe is a new principle whereby simultaneous recognition of multiple independent Tn-glycopeptide antigens on a cancer cell makes tumor escape due to antigen loss unlikely.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Adoptive Transfer , Animals , Antigens, Tumor-Associated, Carbohydrate/immunology , Cell Line , Glycosylation , Humans , Membrane Glycoproteins/immunology , Mice , Neoplasms/pathology
9.
Protein Sci ; 28(9): 1567-1581, 2019 09.
Article in English | MEDLINE | ID: mdl-31276610

ABSTRACT

In this article, we consider the role of heterogeneous nucleation in ß-amyloid aggregation. Heterogeneous nucleation is more common and occurs at lower levels of supersaturation than homogeneous nucleation. The nucleation period is also the stage at which most of the polymorphism of amyloids arises, this being one of the defining features of amyloids. We focus on several well-known heterogeneous nucleators of ß-amyloid, including lipid surfaces, especially those enriched in gangliosides and cholesterol, and divalent metal ions. These two broad classes of nucleators affect ß-amyloid particularly in light of the amphiphilicity of these peptides: the N-terminal region, which is largely polar and charged, contains the metal binding site, whereas the C-terminal region is aliphatic and is important in lipid binding. Notably, these two classes of nucleators can interact cooperatively, aggregation begetting greater aggregation.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Membrane Lipids/metabolism , Metals/metabolism , Animals , Binding Sites , Humans , Protein Aggregates , Protein Domains
10.
Protein Expr Purif ; 162: 72-82, 2019 10.
Article in English | MEDLINE | ID: mdl-31022450

ABSTRACT

We present a straightforward, versatile method for expressing and purifying ß-amyloid (Aß40) and transmembrane peptides derived from ß-amyloid precursor protein (Aß55). In principle, these methods should be applicable to other types of strongly aggregating peptides. We start with a DNA plasmid encoding a HexaHis tag with a flexible, hydrophilic linker sequence, followed by a cleavage site, and then Aß peptides. The HexaHis tag rather than a protein fusion partner (e.g., GST) obviates the need for a folded protein in affinity purification. Second, we present two cleavage methods, using either Factor Xa or BNPS-Skatole. Although the latter procedure requires subsequent reduction of the product, we describe methods for minimizing side reactions. Because the use of BNPS-Skatole obviates the need for a folded protein in the cleavage reaction, it is compatible with harsh conditions (e.g., inclusion of detergents and denaturants) needed to solubilize the fusion proteins; such conditions tend to inactivate Factor Xa. Finally, we also describe purification strategies for Aß40 and Aß55 using FPLC and/or reverse phase HPLC. Yields of peptide after these BNPS-Skatole cleavage and peptide reduction, though subquantitative, greatly exceed those obtained using Factor Xa cleavage, as the reaction of BNPS-Skatole is insensitive to the presence of detergents and denaturants, and therefore can be used to produce highly aggregative and low solubility peptides such as Aß55. Trp is a low abundance amino acid in proteins generally, and for peptides like Aß55, and other transmembane peptides lacking Trp in relevant positions, this cleavage method remains a useful option.


Subject(s)
Amyloid beta-Peptides/chemistry , Biochemistry/methods , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/isolation & purification , Amyloid beta-Peptides/metabolism , Biocatalysis , Chromatography, Affinity , Chromatography, High Pressure Liquid , Factor Xa/chemistry , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility
11.
Nat Med ; 25(4): 690-700, 2019 04.
Article in English | MEDLINE | ID: mdl-30936544

ABSTRACT

Epithelial barrier loss is a driver of intestinal and systemic diseases. Myosin light chain kinase (MLCK) is a key effector of barrier dysfunction and a potential therapeutic target, but enzymatic inhibition has unacceptable toxicity. Here, we show that a unique domain within the MLCK splice variant MLCK1 directs perijunctional actomyosin ring (PAMR) recruitment. Using the domain structure and multiple screens, we identify a domain-binding small molecule (divertin) that blocks MLCK1 recruitment without inhibiting enzymatic function. Divertin blocks acute, tumor necrosis factor (TNF)-induced MLCK1 recruitment as well as downstream myosin light chain (MLC) phosphorylation, barrier loss, and diarrhea in vitro and in vivo. Divertin corrects barrier dysfunction and prevents disease development and progression in experimental inflammatory bowel disease. Beyond applications of divertin in gastrointestinal disease, this general approach to enzymatic inhibition by preventing access to specific subcellular sites provides a new paradigm for safely and precisely targeting individual properties of enzymes with multiple functions.


Subject(s)
Homeostasis , Intestinal Mucosa/metabolism , Intracellular Space/enzymology , Myosin-Light-Chain Kinase/metabolism , Actomyosin/metabolism , Animals , Caco-2 Cells , Chronic Disease , Homeostasis/drug effects , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Jejunum/drug effects , Jejunum/metabolism , Jejunum/pathology , Mice , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/chemistry , Phosphorylation/drug effects , Protein Domains , Small Molecule Libraries/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Macromol Biosci ; 17(2)2017 02.
Article in English | MEDLINE | ID: mdl-27615730

ABSTRACT

Aggregation of denatured or unfolded proteins establishes a large energy barrier to spontaneous recovery of protein form and function following traumatic injury, tissue cryopreservation, and biopharmaceutical storage. Some tissues utilize small heat shock proteins (sHSPs) to prevent irreversible aggregation, which allows more complex processes to refold or remove the unfolded proteins. It is postulated that large, amphiphilic, and biocompatible block copolymers can mimic sHSP function. Reduced and denatured hen egg white lysozyme (HEWL) is used as a model aggregating protein. The poloxamine T1107 prevents aggregation of HEWL at 37 °C by three complimentary measures. Structural analysis of denatured HEWL reveals a partially folded conformation with preserved or promoted beta-sheet structures only in the presence of T1107. The physical association of T1107 with denatured HEWL, and the ability to prevent aggregation, is linked to the critical micelle temperature of the polymer. The results suggest that T1107, or a similar amphiphilic block copolymer, can find use as a synthetic chaperone to augment the innate molecular repair mechanisms of natural cells.


Subject(s)
Muramidase/chemistry , Protein Aggregates , Protein Denaturation , Surface-Active Agents/chemistry , Animals , Cattle , Circular Dichroism , Dithiothreitol/pharmacology , Dynamic Light Scattering , Ethylenediamines/chemistry , Oxidation-Reduction , Particle Size , Protein Denaturation/drug effects , Spectrometry, Fluorescence , Tryptophan/metabolism
13.
J Pept Sci ; 22(5): 368-73, 2016 May.
Article in English | MEDLINE | ID: mdl-27114096

ABSTRACT

Current evidence suggests that oligomers of the amyloid-ß (Aß) peptide are involved in the cellular toxicity of Alzheimer's disease, yet their biophysical characterization remains difficult because of lack of experimental control over the aggregation process under relevant physiologic conditions. Here, we show that modification of the Aß peptide backbone at Gly29 allows for the formation of oligomers but inhibits fibril formation at physiologic temperature and pH. Our results suggest that the putative bend region in Aß is important for higher-order aggregate formation. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid/chemistry , Peptide Fragments/chemical synthesis , Amino Acid Sequence , Glycine/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Mutation , Peptide Fragments/chemistry , Temperature
14.
J Immunol ; 196(4): 1933-42, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26755823

ABSTRACT

CD1c is abundantly expressed on human dendritic cells (DC) and B cells, where it binds and displays lipid Ags to T cells. In this study, we report that CD1c tetramers carrying Mycobacterium tuberculosis phosphomycoketide bind γδ TCRs. An unbiased method of ligand-based TCR selection detects interactions only with Vδ1(+) TCRs, and mutational analyses demonstrate a role of the Vδ1 domain during recognition. These results strengthen evidence for a role of CD1c in the γδ T cell response, providing biophysical evidence for CD1c-γδ TCR interactions and a named foreign Ag. Surprisingly, TCRs also bind CD1c complexes formed with diverse lipids such as lysophosphatidylcholine, sulfatide, or mannosyl-phosophomycoketide, but not lipopeptide ligands. Dissection of TCR interactions with CD1c carrying foreign Ags, permissive ligands, and nonpermissive lipid ligands clarifies the molecular basis of the frequently observed but poorly understood phenomenon of mixed self- and foreign Ag reactivity in the CD1 system.


Subject(s)
Antigen Presentation/immunology , Antigens, CD1/immunology , Glycoproteins/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Flow Cytometry , Humans , Ligands , Polymerase Chain Reaction , Transduction, Genetic
15.
Methods Mol Biol ; 1345: 299-312, 2016.
Article in English | MEDLINE | ID: mdl-26453221

ABSTRACT

Seeding of amyloid fibrils into fresh solutions of the same peptide or protein in disaggregated form leads to the formation of replicate fibrils, with close structural similarity or identity to the original fibrillar seeds. Here we describe procedures for isolating fibrils composed mainly of ß-amyloid (Aß) from human brain and from leptomeninges, a source of cerebral blood vessels, for investigating Alzheimer's disease and cerebral amyloid angiopathy. We also describe methods for seeding isotopically labeled, disaggregated Aß peptide solutions for study using solid-state NMR and other techniques. These methods should be applicable to other types of amyloid fibrils, to Aß fibrils from mice or other species, tissues other than brain, and to some non-fibrillar aggregates. These procedures allow for the examination of authentic amyloid fibrils and other protein aggregates from biological tissues without the need for labeling the tissue.


Subject(s)
Amyloid/chemistry , Isotope Labeling/methods , Magnetic Resonance Spectroscopy/methods , Meninges/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid/ultrastructure , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Brain/metabolism , Brain/ultrastructure , Brain Chemistry , Humans , Meninges/metabolism , Meninges/ultrastructure , Mice , Microscopy, Atomic Force/methods
16.
Biochemistry ; 54(26): 4050-62, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26070092

ABSTRACT

Aggregation of proteins to fiberlike aggregates often involves a transformation of native monomers to ß-sheet-rich oligomers. This general observation underestimates the importance of α-helical segments in the aggregation cascade. Here, using a combination of experimental techniques and accelerated molecular dynamics simulations, we investigate the aggregation of a 43-residue, apolipoprotein A-I mimetic peptide and its E21Q and D26N mutants. Our study indicates a strong propensity of helical segments not to adopt cross-ß-fibrils. The helix-turn-helix monomeric conformation of the peptides is preserved in the mature fibrils. Furthermore, we reveal opposite effects of mutations on and near the turn region in the self-assembly of these peptides. We show that the E21-R24 salt bridge is a major contributor to helix-turn-helix folding, subsequently leading to abundant fibril formation. On the other hand, the K19-D26 interaction is not required to fold the native helix-turn-helix peptide. However, removal of the charged D26 residue decreases the stability of the helix-turn-helix monomer and consequently reduces the level of aggregation. Finally, we provide a more refined assembly model for the helix-turn-helix peptides from apolipoprotein A-I based on the parallel stacking of helix-turn-helix dimers.


Subject(s)
Amyloid/chemistry , Apolipoprotein A-I/chemistry , Peptides/chemistry , Protein Aggregates , Amino Acid Sequence , Amyloid/genetics , Amyloid/ultrastructure , Apolipoprotein A-I/genetics , Apolipoprotein A-I/ultrastructure , Humans , Models, Molecular , Molecular Sequence Data , Point Mutation , Protein Structure, Secondary
17.
J Biol Chem ; 289(49): 33971-83, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25336651

ABSTRACT

P311, a conserved 8-kDa intracellular protein expressed in brain, smooth muscle, regenerating tissues, and malignant glioblastomas, represents the first documented stimulator of TGF-ß1-3 translation in vitro and in vivo. Here we initiated efforts to define the mechanism underlying P311 function. PONDR® (Predictor Of Naturally Disordered Regions) analysis suggested and CD confirmed that P311 is an intrinsically disordered protein, therefore requiring an interacting partner to acquire tertiary structure and function. Immunoprecipitation coupled with mass spectroscopy identified eIF3 subunit b (eIF3b) as a novel P311 binding partner. Immunohistochemical colocalization, GST pulldown, and surface plasmon resonance studies revealed that P311-eIF3b interaction is direct and has a Kd of 1.26 µm. Binding sites were mapped to the non-canonical RNA recognition motif of eIF3b and a central 11-amino acid-long region of P311, here referred to as eIF3b binding motif. Disruption of P311-eIF3b binding inhibited translation of TGF-ß1, 2, and 3, as indicated by luciferase reporter assays, polysome fractionation studies, and Western blot analysis. RNA precipitation assays after UV cross-linking and RNA-protein EMSA demonstrated that P311 binds directly to TGF-ß 5'UTRs mRNAs through a previously unidentified RNA recognition motif-like motif. Our results demonstrate that P311 is a novel RNA-binding protein that, by interacting with TGF-ßs 5'UTRs and eIF3b, stimulates the translation of TGF-ß1, 2, and 3.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins/metabolism , Protein Biosynthesis , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/metabolism , 5' Untranslated Regions , Amino Acid Sequence , Animals , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NIH 3T3 Cells , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Protein Binding , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta2/chemistry , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics
18.
Structure ; 22(7): 996-1007, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24931469

ABSTRACT

Human presequence protease (hPreP) is an M16 metalloprotease localized in mitochondria. There, hPreP facilitates proteostasis by utilizing an ∼13,300-Å(3) catalytic chamber to degrade a diverse array of potentially toxic peptides, including mitochondrial presequences and ß-amyloid (Aß), the latter of which contributes to Alzheimer disease pathogenesis. Here, we report crystal structures for hPreP alone and in complex with Aß, which show that hPreP uses size exclusion and charge complementation for substrate recognition. These structures also reveal hPreP-specific features that permit a diverse array of peptides, with distinct distributions of charged and hydrophobic residues, to be specifically captured, cleaved, and have their amyloidogenic features destroyed. SAXS analysis demonstrates that hPreP in solution exists in dynamic equilibrium between closed and open states, with the former being preferred. Furthermore, Aß binding induces the closed state and hPreP dimerization. Together, these data reveal the molecular basis for flexible yet specific substrate recognition and degradation by hPreP.


Subject(s)
Mitochondrial Proteins/chemistry , Peptides/chemistry , Serine Endopeptidases/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Crystallography, X-Ray , Humans , Kinetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , Scattering, Small Angle , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Substrate Specificity , X-Ray Diffraction
19.
J Am Chem Soc ; 136(27): 9619-26, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-24926593

ABSTRACT

The 99 amino acid C-terminal fragment of amyloid precursor protein (C99), consisting of a single transmembrane (TM) helix, is known to form homodimers. Homodimers can be processed by γ-secretase to produce amyloid-ß (Aß) protein, which is implicated in Alzheimer's disease (AD). While knowledge of the structure of C99 homodimers is of great importance, experimental NMR studies and simulations have produced varying structural models, including right-handed and left-handed coiled-coils. In order to investigate the structure of this critical protein complex, simulations of the C99(15-55) homodimer in POPC membrane bilayer and DPC surfactant micelle environments were performed using a multiscale approach that blends atomistic and coarse-grained models. The C99(15-55) homodimer adopts a dominant right-handed coiled-coil topology consisting of three characteristic structural states in a bilayer, only one of which is dominant in the micelle. Our structural study, which provides a self-consistent framework for understanding a number of experiments, shows that the energy landscape of the C99 homodimer supports a variety of slowly interconverting structural states. The relative importance of any given state can be modulated through environmental selection realized by altering the membrane or micelle characteristics.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Micelles , Models, Molecular , Protein Conformation
20.
J Am Chem Soc ; 136(3): 854-7, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24364734

ABSTRACT

The amyloid ß (Aß) peptide associated with Alzheimer's disease results from processing of the amyloid precursor protein (APP) by secretases. Cleavage of APP by ß-secretase produces a 99 amino acid C-terminal fragment of APP (C99) consisting of a single transmembrane (TM) helix. Simulations of C99 congeners and structural studies of C99 in surfactant micelles and lipid vesicles have shown that a key peptide structural motif is a prominent "GG kink," centered at two glycines dividing the TM helix. The flexibility of the GG kink is important in the processing of C99 by γ-secretase. We performed multiscale simulations of C99(15-55) in a DPC surfactant micelle and POPC lipid bilayer in order to elucidate the role of membrane surface curvature in modulating the peptide structure. C99(15-55) in a DPC surfactant micelle possesses a "GG kink," in the TM domain near the dynamic hinge located at G37/G38. Such a kink is not observed in C99(15-55) in a POPC lipid bilayer. Intramolecular interaction between the extracellular and TM domains of C99(15-55) is enhanced in the micelle environment, influencing helical stability, TM helix extension, exposure to water, and depth of insertion in the lipophilic region. Our results show that the fluctuations of the structural ensemble of APP are strongly influenced by membrane surface curvature.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Cell Membrane/chemistry , Models, Molecular , Amino Acid Sequence , Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Molecular Sequence Data , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...