Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Elife ; 102021 12 29.
Article in English | MEDLINE | ID: mdl-34964715

ABSTRACT

Mechanosensory transduction (MT), the conversion of mechanical stimuli into electrical signals, underpins hearing and balance and is carried out within hair cells in the inner ear. Hair cells harbor actin-filled stereocilia, arranged in rows of descending heights, where the tips of stereocilia are connected to their taller neighbors by a filament composed of protocadherin 15 (PCDH15) and cadherin 23 (CDH23), deemed the 'tip link.' Tension exerted on the tip link opens an ion channel at the tip of the shorter stereocilia, thus converting mechanical force into an electrical signal. While biochemical and structural studies have provided insights into the molecular composition and structure of isolated portions of the tip link, the architecture, location, and conformational states of intact tip links, on stereocilia, remains unknown. Here, we report in situ cryo-electron microscopy imaging of the tip link in mouse stereocilia. We observe individual PCDH15 molecules at the tip and shaft of stereocilia and determine their stoichiometry, conformational heterogeneity, and their complexes with other filamentous proteins, perhaps including CDH23. The PCDH15 complexes occur in clusters, frequently with more than one copy of PCDH15 at the tip of stereocilia, suggesting that tip links might consist of more than one copy of PCDH15 complexes and, by extension, might include multiple MT complexes.


Subject(s)
Cadherin Related Proteins/chemistry , Protein Precursors/chemistry , Stereocilia/ultrastructure , Animals , Cadherin Related Proteins/ultrastructure , Cryoelectron Microscopy , Mice , Molecular Conformation , Molecular Structure , Protein Precursors/ultrastructure
2.
J Dermatol Sci ; 100(1): 39-49, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32893105

ABSTRACT

BACKGROUND: Profilaggrin belongs to the S100 fused-type protein family expressed in keratinocytes and is important for skin barrier integrity. Its N-terminus contains an S100 ("A") domain and a unique "B" domain with a nuclear localization sequence. OBJECTIVE: To determine whether profilaggrin B domain cooperates with the S100 domain to bind macromolecules. To characterize the biochemical and structural properties of the profilaggrin N-terminal "AB" domain and compare it to other S100 fused-type proteins. METHODS: We used biochemical (protease protection, light scattering, fluorescence spectroscopy, pull-down assays) and computational techniques (sequence analysis, molecular modeling with crystallographic structures) to examine human profilaggrin and S100 fused-type proteins. RESULTS: Comparing profilaggrin S100 crystal structure with models of the other S100 fused-type proteins demonstrated each has a unique chemical composition of solvent accessible surface around the hydrophobic binding pocket. S100 fused-type proteins exhibit higher pocket hydrophobicity than soluble S100 proteins. The inter-EF-hand linker in S100 fused-type proteins contains conserved hydrophobic residues involved in binding substrates. Profilaggrin B domain cooperates with the S100 domain to bind annexin II and keratin intermediate filaments in a calcium-dependent manner using exposed cationic surface. Using molecular modeling we demonstrate profilaggrin B domain likely interacts with annexin II domains I and II. Steric clash analysis shows annexin II N-terminal peptide is favored to bind profilaggrin among S100 fused-type proteins. CONCLUSION: The N-terminal S100 and B domains of profilaggrin cooperate to bind substrate molecules in granular layer keratinocytes to provide epidermal barrier functions.


Subject(s)
Intermediate Filament Proteins/ultrastructure , Protein Precursors/ultrastructure , S100 Proteins/metabolism , Amino Acid Sequence , Annexin A2/genetics , Annexin A2/isolation & purification , Annexin A2/metabolism , Annexin A2/ultrastructure , Binding Sites/genetics , Crystallography, X-Ray , Filaggrin Proteins , Humans , Hydrophobic and Hydrophilic Interactions , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/isolation & purification , Intermediate Filament Proteins/metabolism , Intermediate Filaments/metabolism , Keratinocytes , Keratins/genetics , Keratins/isolation & purification , Keratins/metabolism , Keratins/ultrastructure , Molecular Docking Simulation , Mutation , Protein Binding/genetics , Protein Conformation, alpha-Helical/genetics , Protein Domains/genetics , Protein Precursors/genetics , Protein Precursors/isolation & purification , Protein Precursors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
3.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070639

ABSTRACT

Hearing and balance involve the transduction of mechanical stimuli into electrical signals by deflection of bundles of stereocilia linked together by protocadherin 15 (PCDH15) and cadherin 23 'tip links'. PCDH15 transduces tip link tension into opening of a mechano-electrical transduction (MET) ion channel. PCDH15 also interacts with LHFPL5, a candidate subunit of the MET channel. Here we illuminate the PCDH15-LHFPL5 structure, showing how the complex is composed of PCDH15 and LHFPL5 subunit pairs related by a 2-fold axis. The extracellular cadherin domains define a mobile tether coupled to a rigid, 2-fold symmetric 'collar' proximal to the membrane bilayer. LHFPL5 forms extensive interactions with the PCDH15 transmembrane helices and stabilizes the overall PCDH15-LHFPL5 assembly. Our studies illuminate the architecture of the PCDH15-LHFPL5 complex, localize mutations associated with deafness, and shed new light on how forces in the PCDH15 tether may be transduced into the stereocilia membrane.


Subject(s)
Cadherins/chemistry , Cadherins/metabolism , Membrane Proteins/metabolism , Protein Precursors/chemistry , Protein Precursors/metabolism , Stereocilia/metabolism , Amino Acid Sequence , Animals , Cadherin Related Proteins , Cadherins/ultrastructure , HEK293 Cells , Humans , Imaging, Three-Dimensional , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Mice , Models, Molecular , Protein Multimerization , Protein Precursors/ultrastructure , Sf9 Cells
4.
Sci Rep ; 8(1): 3837, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497069

ABSTRACT

The hepatitis B virus (HBV) infection is a critical health problem worldwide, and HBV preS1 is an important biomarker for monitoring HBV infection. Previously, we found that a murine monoclonal antibody, mAb-D8, targets the preS1 (aa91-107) fragment of HBV. To improve its performance, we prepared the single-chain variable region of mAb-D8 (scFvD8) and constructed the three-dimensional structure of the scFvD8-preS1 (aa91-107) complex by computer modelling. The affinity of scFvD8 was markedly increased by the introduction of mutations L96Tyr to Ser and H98Asp to Ser. Furthermore, a highly sensitive immunosensor was designed based on a proximity-dependent hybridization strategy in which the preS1 antigen competitively reacts with an antibody labelled with DNA, resulting in decreased proximity-dependent hybridization and increased electrochemical signal from the Fc fragment, which can be used for the quantisation of preS1. The results showed a wide detection range from 1 pM to 50 pM with a detection limit of 0.1 pM. The sensitivity and specificity of this immunosensor in clinical serum samples were 100% and 96%, respectively. This study provides a novel system based on proximity-dependent hybridization and the scFv antibody fragment for the rapid quantisation of antigens of interest with a high sensitivity.


Subject(s)
Hepatitis B Antibodies/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/ultrastructure , Protein Precursors/immunology , Protein Precursors/ultrastructure , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Hepatitis B/virology , Hepatitis B Surface Antigens/blood , Hepatitis B virus/genetics , Humans , Hybridomas/immunology , Mice , Nucleic Acid Hybridization/methods , Protein Precursors/genetics , Single-Chain Antibodies/genetics
5.
PLoS One ; 12(7): e0181799, 2017.
Article in English | MEDLINE | ID: mdl-28746363

ABSTRACT

Little structural information is available so far on amyloid fibrils consisting of immunoglobulin light chains. It is not understood which features of the primary sequence of the protein result in fibril formation. We report here MAS solid-state NMR studies to identify the structured core of κ-type variable domain light chain fibrils. The core contains residues of the CDR2 and the ß-strands D, E, F and G of the native immunoglobulin fold. The assigned core region of the fibril is distinct in comparison to the core identified in a previous solid-state NMR study on AL-09 by Piehl at. al, suggesting that VL fibrils can adopt different topologies. In addition, we investigated a soluble oligomeric intermediate state, previously termed the alternatively folded state (AFS), using NMR and FTIR spectroscopy. The NMR oligomer spectra display a high degree of similarity when compared to the fibril spectra, indicating a high structural similarity of the two aggregation states. Based on comparison to the native state NMR chemical shifts, we suggest that fibril formation via domain-swapping seems unlikely. Moreover, we used our results to test the quality of different amyloid prediction algorithms.


Subject(s)
Amyloid/chemistry , Immunoglobulin Light Chains/chemistry , Protein Multimerization , Protein Precursors/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Models, Molecular , Mutation , Protein Conformation , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Spectroscopy, Fourier Transform Infrared
6.
Protein Expr Purif ; 100: 10-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24810910

ABSTRACT

The HIV-1 Gag precursor protein, Pr55(Gag), is a multi-domain polyprotein that drives HIV-1 assembly. The morphological features of HIV-1 suggested Pr55(Gag) assumes a variety of different conformations during virion assembly and maturation, yet structural determination of HIV-1 Pr55(Gag) has not been possible due to an inability to express and to isolate large amounts of full-length recombinant Pr55(Gag) for biophysical and biochemical analyses. This challenge is further complicated by HIV-1 Gag's natural propensity to multimerize for the formation of viral particle (with ∼2500 Gag molecules per virion), and this has led Pr55(Gag) to aggregate and be expressed as inclusion bodies in a number of in vitro protein expression systems. This study reported the production of a recombinant form of HIV-1 Pr55(Gag) using a bacterial heterologous expression system. Recombinant HIV-1 Pr55(Gag) was expressed with a C-terminal His×6 tag, and purified using a combination of immobilized metal affinity chromatography and size exclusion chromatography. This procedure resulted in the production of milligram quantities of high purity HIV-1 Pr55(Gag) that has a mobility that resembles a trimer in solution using size exclusion chromatography analysis. The high quantity and purity of the full length HIV Gag will be suitable for structural and functional studies to further understand the process of viral assembly, maturation and the development of inhibitors to interfere with the process.


Subject(s)
Escherichia coli/genetics , HIV Infections/virology , HIV-1/genetics , Protein Precursors/genetics , Protein Precursors/isolation & purification , Amino Acid Sequence , Base Sequence , Chromatography, Affinity/methods , Gene Expression , HIV-1/chemistry , Humans , Metals/chemistry , Molecular Sequence Data , Plasmids/genetics , Protein Precursors/chemistry , Protein Precursors/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/ultrastructure , Solubility , Transformation, Bacterial
7.
Sci China Life Sci ; 53(9): 1049-56, 2010 Sep.
Article in English | MEDLINE | ID: mdl-21104364

ABSTRACT

SecA, the ATPase of Sec translocase, mediates the post-translational translocation of preprotein through the protein-conducting channel SecYEG in the bacterial inner membrane. Here we report the structures of Escherichia coli Sec intermediates during preprotein translocation as visualized by electron microscopy to probe the oligomeric states of SecA during this process. We found that the translocase holoenzyme is symmetrically assembled by SecA and SecYEG on proteoliposomes, whereas the translocation intermediate 31 (I(31)) becomes asymmetric because of the presence of preprotein. Moreover, SecA is a dimer in these two translocation complexes. This work also shows surface topological changes in the components of translocation intermediates by immunogold labeling. The channel entry for preprotein translocation was found at the center of the I(31) structures. Our results indicate that the presence of preprotein introduces asymmetry into translocation intermediates, while SecA remains dimeric during the translocation process.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/ultrastructure , Protein Structure, Quaternary , Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Dimerization , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Immunohistochemistry , Membrane Transport Proteins/metabolism , Protein Precursors/chemistry , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Proteolipids/chemistry , SEC Translocation Channels , SecA Proteins
8.
Sci Signal ; 2(85): pt5, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706872

ABSTRACT

Mechanotransduction, the conversion of mechanical force into an electrochemical signal, allows living organisms to detect touch, hear, register movement and gravity, and sense changes in cell volume and shape. Hair cells in the vertebrate inner ear are mechanoreceptor cells specialized for the detection of sound and head movement. Each hair cell contains, at the apical surface, rows of stereocilia that are connected by extracellular filaments to form an exquisitely organized bundle. Mechanotransduction channels, localized near the tips of the stereocilia, are gated by the gating spring, an elastic element that is stretched upon stereocilia deflection and mediates rapid channel opening. Components of the mechanotransduction machinery in hair cells have been identified and several are encoded by genes linked to deafness in humans, which indicates that defects in the mechanotransduction machinery are the underlying cause of some forms of hearing impairment.


Subject(s)
Hair Cells, Auditory/physiology , Mechanotransduction, Cellular/physiology , Animals , Cadherin Related Proteins , Cadherins/physiology , Cadherins/ultrastructure , Caenorhabditis elegans/physiology , Carrier Proteins/physiology , Carrier Proteins/ultrastructure , Cell Cycle Proteins , Cytoskeletal Proteins , Hair Cells, Auditory/ultrastructure , Hearing/physiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Ion Channel Gating/physiology , Mechanoreceptors/physiology , Mice , Mice, Knockout , Mice, Neurologic Mutants , Microscopy, Immunoelectron , Protein Interaction Mapping , Protein Precursors/physiology , Protein Precursors/ultrastructure , Touch/physiology
9.
Biochemistry ; 48(17): 3778-86, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19281242

ABSTRACT

Amyloid fibrils are found in approximately 25 different diseases, including Alzheimer's disease. Lung surfactant protein C (SP-C) forms fibrils in association with pulmonary disease. It was recently found that the C-terminal domain of proSP-C (CTC), which is localized to the endoplasmic reticulum (ER) lumen, protects the transmembrane (TM) part of (pro)SP-C from aggregation into amyloid until it has a folded into an alpha-helix. CTC appears to have a more general anti-amyloid effect by also acting on TM regions of other proteins. Here we investigate interactions of CTC with the amyloid beta-peptide (Abeta) associated with Alzheimer's disease and medin, a peptide that forms fibrils in the most common form of human amyloid. CTC prevents fibril formation in Abeta and medin and forms a complex with Abeta oligomers, as judged by size-exclusion chromatography and electrospray ionization mass spectrometry. These data suggest that CTC functions as a chaperone that acts preferentially against unfolded TM segments and structural motifs found during amyloid fibril formation, a mechanism that may be exploited in forming a basis for future anti-amyloid therapy.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Milk Proteins/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Protein Precursors/physiology , Pulmonary Surfactant-Associated Protein C/physiology , Amino Acid Sequence , Amyloid beta-Peptides/ultrastructure , Antigens, Surface/ultrastructure , Humans , Milk Proteins/ultrastructure , Molecular Chaperones/chemistry , Molecular Chaperones/physiology , Molecular Chaperones/ultrastructure , Molecular Sequence Data , Peptide Fragments/ultrastructure , Protein Folding , Protein Precursors/ultrastructure , Protein Structure, Tertiary/physiology , Pulmonary Surfactant-Associated Protein C/chemistry , Pulmonary Surfactant-Associated Protein C/ultrastructure
10.
J Comp Neurol ; 512(3): 419-31, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19009591

ABSTRACT

The dynorphin (DYN)-kappa opioid receptor (kappaOR) system has been implicated in stress modulation, depression, and relapse to drug-seeking behaviors. Previous anatomical and physiological data have indicated that the noradrenergic nucleus locus coeruleus (LC) is one site at which DYN may contribute to these effects. Using light microscopy, immunofluorescence, and electron microscopy, the present study investigated the cellular substrates for pre- and postsynaptic interactions of kappaOR in the LC. Dual immunocytochemical labeling for kappaOR and tyrosine hydroxylase (TH) or kappaOR and preprodynorphin (ppDYN) was examined in the same section of tissue. Light microscopic analysis revealed prominent kappaOR immunoreactivity in the nuclear core of the LC and in the peri-coerulear region where noradrenergic dendrites extend. Fluorescence and electron microscopy revealed kappaOR immunoreactivity within TH-immunoreactive somata and dendrites in the LC as well as localized to ppDYN-immunoreactive processes. In sections processed for kappaOR and TH, approximately 29% (200/688) of the kappaOR-containing axon terminals identified targeted TH-containing profiles. Approximately 49% (98/200) of the kappaOR-labeled axon terminals formed asymmetric synapses with TH-labeled dendrites. Sections processed for kappaOR and ppDYN showed that, of the axon terminals exhibiting kappaOR, 47% (223/477) also exhibited ppDYN. These findings indicate that kappaORs are poised to modulate LC activity by their localization to somata and dendrites. Furthermore, kappaORs are strategically localized to presynaptically modulate DYN afferent input to catecholamine-containing neurons in the LC. These data add to the growing literature showing that kappaORs can modulate diverse afferent signaling to the LC.


Subject(s)
Locus Coeruleus/cytology , Receptors, Opioid, kappa/metabolism , Animals , Dynorphins/metabolism , Locus Coeruleus/metabolism , Male , Microscopy, Immunoelectron , Neurons/metabolism , Neurons/ultrastructure , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/ultrastructure , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/ultrastructure
11.
J Cell Biochem ; 102(5): 1149-59, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17654502

ABSTRACT

Pre-lamin A undergoes subsequent steps of post-translational modification at its C-terminus, including farnesylation, methylation, and cleavage by ZMPSTE24 metalloprotease. Here, we show that accumulation of different intermediates of pre-lamin A processing in nuclei, induced by expression of mutated pre-lamin A, differentially affected chromatin organization in human fibroblasts. Unprocessed (non-farnesylated) pre-lamin A accumulated in intranuclear foci, caused the redistribution of LAP2alpha and of the heterochromatin markers HP1alpha and trimethyl-K9-histone 3, and triggered heterochromatin localization in the nuclear interior. In contrast, the farnesylated and carboxymethylated lamin A precursor accumulated at the nuclear periphery and caused loss of heterochromatin markers and Lap2alpha in enlarged nuclei. Interestingly, pre-lamin A bound both HP1alpha and LAP2alpha in vivo, but the farnesylated form showed reduced affinity for HP1alpha. Our data show a link between pre-lamin A processing and heterochromatin remodeling and have major implications for understanding molecular mechanisms of human diseases linked to mutations in lamins.


Subject(s)
Heterochromatin/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Biopsy, Needle , Cell Nucleus/metabolism , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/ultrastructure , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Dermatologic Surgical Procedures , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fluorescent Antibody Technique, Indirect , Heterochromatin/genetics , Heterochromatin/ultrastructure , Humans , Lamin Type A , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/ultrastructure , Precipitin Tests , Protein Precursors/genetics , Protein Precursors/ultrastructure , Skin/cytology , Transfection
12.
FEBS Lett ; 581(8): 1635-8, 2007 Apr 17.
Article in English | MEDLINE | ID: mdl-17399708

ABSTRACT

Recent studies have led us to suppose that synergistic action of multiple solute cofactors could play substantial roles in amyloid-type fibrillogenesis of pathogenic polypeptides. To support this view, we performed aggregation experiments of human islet amyloid polypeptide (IAPP) in media containing both polyanions and non-polar solvents. The results demonstrated that the fibrillation at sub-micromolar IAPP occurred only when polyanionic and non-polar solutes coexist. A simple sum of two independent cofactor's effects could not account for the synergistic action. We propose that this synergy of polyanionic and nonpolar milieus could substantially modify the amyloidgenesis in the human body.


Subject(s)
Amyloid/ultrastructure , Polymers/chemistry , Protein Precursors/ultrastructure , Alcohols/chemistry , Amyloid/chemistry , Glycosaminoglycans/chemistry , Heparin/chemistry , Humans , Islet Amyloid Polypeptide , Polyelectrolytes , Polytetrafluoroethylene/chemistry , Protein Precursors/chemistry , Solutions/chemistry
13.
Biopolymers ; 88(2): 239-52, 2007.
Article in English | MEDLINE | ID: mdl-17206626

ABSTRACT

The study of conformational transitions of peptides has obtained considerable attention recently because of their importance as a molecular key event in a variety of degenerative diseases. However, the study of peptide self-assembly into beta-sheets and amyloid beta (Abeta) fibrils is strongly hampered by their difficult synthetic access and low solubility. We have recently developed a new concept termed switch-peptides that allows the controlled onset of polypeptide folding and misfolding at physiologic conditions. As a major feature, the folding process is initiated by chemically or enzyme triggered O,N-acyl migration in flexible and soluble folding precursors containing Ser- or Thr-derived switch (S)-elements. The elaborated methodologies are exemplified for the in situ conversion of NPY- and Cyclosporine A-derived prodrugs, as well as for the onset and reversal of alpha and beta conformational transitions in Abeta peptides. In combining orthogonally addressable switch-elements, the consecutive switching on of S-elements gives new insights into the role of individual peptide segments (hot spots) in early processes of polypeptide self-assembly and fibrillogenesis. Finally, the well-known secondary structure disrupting effect of pseudoprolines (PsiPro) is explored for its use as a building block (S-element) in switch-peptides. To this end, synthetic strategies are described, allowing for the preparation of PsiPro-containing folding precursors, exhibiting flexible random-coil conformations devoid of fibril forming propensity. The onset of beta-sheet and fibril formation by restoring the native peptide chain in a single step classify PsiPro-units as the most powerful tool for inhibiting peptide self-assembly, and complement the present methodologies of the switch-concept for the study of fibrillogenesis.


Subject(s)
Amyloid/chemistry , Peptides/chemistry , Amyloid/ultrastructure , Microscopy, Electron , Models, Biological , Models, Molecular , Proline/analogs & derivatives , Proline/chemistry , Protein Folding , Protein Precursors/chemistry , Protein Precursors/ultrastructure , Protein Structure, Secondary , Thiazoles/chemistry
14.
Biochem Biophys Res Commun ; 352(3): 592-7, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17141187

ABSTRACT

We have applied NMR spectroscopy to determine the high-resolution structure of gaegurin 4, a 37-residue antimicrobial peptide from Rana rugosa, under varying hydrophobic conditions. Even in 100% H2O, gaegurin 4 contains a nascent turn near its C-terminal Rana box. Under a more hydrophobic condition it forms two amphipathic helices, one long encompassing residues 2-23 and the other consisting of residues 25-34, similar to what has been observed in cecropin A. Functional implication of the helix-breaking kink at Gly24 in gaegurin 4 was investigated by preparing several analogs. Based upon the current and previous results, we propose a novel seaanemone-like ion pore-forming model for gaegurin 4.


Subject(s)
Cell Membrane/chemistry , Models, Chemical , Models, Molecular , Protein Precursors/chemistry , Protein Precursors/ultrastructure , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Computer Simulation , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Conformation , Solutions
15.
FEBS J ; 273(5): 926-35, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16478467

ABSTRACT

Mature lung surfactant protein C (SP-C) corresponds to residues 24-58 of the 21 kDa proSP-C. A late processing intermediate, SP-Ci, corresponding to residues 12-58 of proSP-C, lacks the surface activity of SP-C, and the SP-Ci alpha-helical structure does not unfold in contrast to the metastable nature of the SP-C helix. The NMR structure of an analogue of SP-Ci, SP-Ci(1-31), with two palmitoylCys replaced by Phe and four Val replaced by Leu, in dodecylphosphocholine micelles and in ethanol shows that its alpha-helix vs. that of SP-C is extended N-terminally. The Arg-Phe part in SP-Ci that is cleaved to generate SP-C is localized in a turn structure, which is followed by a short segment in extended conformation. Circular dichroism spectroscopy of SP-Ci(1-31) in microsomal or surfactant lipids shows a mixture of helical and extended conformation at pH 6, and a shift to more unordered structure at pH 5. Replacement of the N-terminal hexapeptide segment SPPDYS (known to constitute a signal in intracellular targeting) of SP-Ci with AAAAAA results in a peptide that is mainly unstructured, independent of pH, in microsomal and surfactant lipids. Addition of a synthetic dodecapeptide, corresponding to the propeptide part of SP-Ci, to mature SP-C results in slower aggregation kinetics and altered amyloid fibril formation, and reduces the surface activity of phospholipid-bound SP-C. These data suggest that the propeptide part of SP-Ci prevents unfolding by locking the N-terminal part of the helix, and that acidic pH results in structural disordering of the region that is proteolytically cleaved to generate SP-C.


Subject(s)
Pulmonary Surfactant-Associated Protein C/chemistry , Animals , Circular Dichroism , Drug Stability , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Micelles , Microscopy, Electron , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Protein Conformation , Protein Precursors/chemistry , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Pulmonary Surfactant-Associated Protein C/metabolism , Pulmonary Surfactant-Associated Protein C/ultrastructure , Rabbits , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Swine
16.
Biophys J ; 89(6): 3701-13, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150975

ABSTRACT

The formation, relative stability, and possible stoichiometries of two (self-)complementary peptide sequences (B and E) designed to form either a parallel homodimeric (B + B) or an antiparallel heterodimeric (B + E) coiled coil have been investigated. Peptide B shows a characteristic coiled coil pattern in circular dichroism spectra at pH 7.4, whereas peptide E is apparently random coiled under these conditions. The peptides are complementary to each other, with peptide E forming a coiled coil when mixed with peptide B. Molecular dynamics simulations show that combinations of B + B and B + E readily form a dimeric coiled coil, whereas E + E does not fall in line with the experimental data. However, the simulations strongly suggest the preferred orientation of the helices in the homodimeric coiled coil is antiparallel, with interactions at the interface quite different to that of the idealized model. In addition, molecular dynamics simulations suggest equilibrium between dimers, trimers, and tetramers of alpha-helices for peptide B.


Subject(s)
Enkephalins/chemistry , Models, Chemical , Models, Molecular , Protein Precursors/chemistry , Binding Sites , Computer Simulation , Dimerization , Drug Stability , Enkephalins/analysis , Kinetics , Multiprotein Complexes/analysis , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Conformation , Protein Precursors/analysis , Protein Precursors/ultrastructure , Protein Structure, Secondary
17.
Biochemistry ; 42(44): 12972-80, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14596612

ABSTRACT

Hsp90 is a molecular chaperone that binds and assists refolding of non-native and/or labile polypeptides and also bind various peptides. However, the rules of how Hsp90 recognizes substrates have not been well characterized. By surface plasmon resonance measurements, a physiologically active peptide, neuropeptide Y (NPY), with a strong binding property to Hsp90 was identified from screening of 38 randomly selected peptide candidates. We showed that the carboxy-terminal fragment of NPY (NPY13-36), which forms an amphipathic alpha-helix structure, preserved the strong binding to Hsp90. Immunoprecipitation and immunoblotting using HeLa cell extracts revealed that newly synthesized NPY precursors bound to Hsp90, suggesting that the in vitro binding experiments identified an interactive peptide in vivo. Proteolytic cleavage of the NPY13-36/Hsp90 complex, as well as binding site analysis using deletion mutants of Hsp90, revealed the NPY binding locus on Hsp90alpha as the 192 amino acid region following the N-terminal domain. By electron microscopic analysis using an anti-Hsp90 antibody against the sequence proximal to the highly charged region, we showed that the Hsp90 dimer bound to NPY13-36 at both ends. Mutation of arginine residues in NPY13-36 to alanine abrogated binding to Hsp90. Our studies indicate that the hinge region after the N-terminal domain of Hsp90 and the positive charges on NPY are important for this interaction.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Neuropeptide Y/chemistry , Neuropeptide Y/metabolism , Amino Acid Sequence , Animals , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/ultrastructure , HeLa Cells , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Protein Binding/genetics , Protein Precursors/chemistry , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Static Electricity , Structure-Activity Relationship , Surface Plasmon Resonance , Swine , Transfection
18.
Adv Protein Chem ; 64: 301-23, 2003.
Article in English | MEDLINE | ID: mdl-13677051

ABSTRACT

Bacteriophage T7 is a double-stranded DNA bacteriophage that has attracted particular interest in studies of gene expression and regulation and of morphogenesis, as well as in biotechnological applications of expression vectors and phage display. We report here studies of T7 capsid assembly by cryoelectron microscopy and image analysis. T7 follows the canonical pathway of first forming a procapsid that converts into the mature capsid, but with some novel variations. The procapsid is a round particle with an icosahedral triangulation number of 7 levo, composed of regular pentamers and elongated hexamers. A singular vertex in the procapsid is occupied by the connector/portal protein, which forms 12-fold and 13-fold rings when overexpressed, of which the 12-mer appears to be the assembly-competent form. This vertex is the site of two symmetry mismatches: between the connector and the surrounding five gp 10 hexamers; and between the connector and the 8-fold cylindrical core mounted on its inner surface. The scaffolding protein, gp9, which is required for assembly, forms nubbin-like protrusions underlying the hexamers but not the pentamers, with no contacts between neighboring gp9 monomers. We propose that gp9 facilitates assembly by binding to gp10 hexamers, locking them into a morphogenically correct conformation. gp9 is expelled as the procapsid matures into the larger, thinner walled, polyhedral capsid. Several lines of evidence implicate the connector vertex as the site at which the maturation transformation is initiated: in vivo, maturation appears to be triggered by DNA packaging whereby the signal may involve interaction of the connector with DNA. In the mature T7 head, the DNA is organized as a tightly wound coaxial spool, with the DNA coiled around the core in at least four and perhaps as many as six concentric shells.


Subject(s)
Bacteriophage T7/chemistry , Bacteriophage T7/physiology , Capsid/chemistry , DNA, Viral/chemistry , Protein Precursors/chemistry , Bacteriophage T7/genetics , Bacteriophage T7/ultrastructure , Capsid/ultrastructure , DNA, Viral/genetics , DNA, Viral/metabolism , DNA, Viral/ultrastructure , Protein Precursors/ultrastructure
19.
J Mol Biol ; 327(1): 1-6, 2003 Mar 14.
Article in English | MEDLINE | ID: mdl-12614603

ABSTRACT

Like other bacteriophages, T7 has a singular vertex that is the site of a symmetry mismatch involving the portal/connector protein, a 12-fold ring at the vertex site which is also a 5-fold axis for the icosahedral capsid. In the mature virion, a 6-fold-symmetric tail extends outwards from the connector. T7 also has a cylindrical "core" that assembles on the inner surface of the connector during procapsid formation, is retained in the mature virion, and is required for infectivity. We have investigated the core structure by cryo-electron microscopy and image analysis of procapsids and find that it observes 8-fold symmetry. Stoichiometry data indicate that its major constituent is an octamer of gp15.


Subject(s)
Bacteriophage T7/chemistry , Bacteriophage T7/ultrastructure , Capsid/chemistry , Capsid/ultrastructure , Protein Precursors/chemistry , Protein Precursors/ultrastructure , Cryoelectron Microscopy , Protein Conformation , Viral Structural Proteins/chemistry , Viral Structural Proteins/ultrastructure , Virion/chemistry , Virion/ultrastructure
20.
Cell Struct Funct ; 27(3): 145-55, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12207045

ABSTRACT

We investigated whether yeast signals could regulate hormone processing in mammalian cells. Chmeric genes coding for the prepro region of yeast alpha-factor and the functional hormone region of anglerfish somatostatin was expressed in rat pituitary GH(3) cells. The nascent prepro-alpha-factor-somatostatin peptides disappeared from cells with a half-life of 30 min, and about 20% of unprocessed precursors remained intracellular after a 2 h chase period. Disappearance of propeptide was insensitive to lysosomotropic agents, but was inhibited at 15 degrees C or 20 degrees C, suggesting that the hybrid propeptides were not degraded in the secretory pathway to the trans Golgi network or in lysosomes. It appeared that while most unprocessed precursors were constitutively secreted into the medium, a small portion were processed at their paired dibasic sites by prohormone-processing enzymes located in trans Golgi network/secretory vesicles, resulting in the production of mature somatostatin peptides. To test this hypothesis, we investigated the processing pattern of two different hybrid precursors: the 52-1 hybrid precursor, which has a Glu-Ala spacer between the prepro region of alpha-factor and somatostatin, and the 58-1 hybrid precursor, which lacks the Glu-Ala spacer. Processing of metabolically labeled hybrid propeptides to smaller somatostatin peptides was assessed by HPLC. When pulse-labeled cells were chased for up to 2 h, 68% of the initially synthesized propeptides were secreted constitutively. About 22% of somatostatin-related products were proteolytically processed to mature somatostatin, of which 38.7% were detected intracellularly after 2 h. From N-terminal peptide sequence determination of somatostatin-related products in GH(3)-52 and GH(3)-58 cells, we found that both hybrid precursors were accurately cleaved at their dibasic amino acid sites. Notably, we also observed that the Glu-Ala spacer sequence was removed from 52-1 hybrid precursors. The latter result strongly suggests that a novel dipeptidyl aminopeptidase activity - a yeast STE13-like enzyme - is present in the post-trans Golgi network compartment of GH(3) cells. The data from these studies indicate that mechanisms which control protein secretion are conserved between yeast and mammalian cells.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Pituitary Gland, Anterior/cytology , Protein Precursors/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Cells, Cultured , Chimera , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Growth Hormone/metabolism , Lipoproteins/biosynthesis , Pheromones , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/ultrastructure , Rats , Saccharomyces cerevisiae Proteins , Somatostatin/biosynthesis , Somatostatin/genetics , Somatostatin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...