Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
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
2.
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
3.
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
4.
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
5.
J Cell Biol ; 107(6 Pt 1): 2045-9, 1988 Dec.
Article in English | MEDLINE | ID: mdl-2848848

ABSTRACT

A purified, artificial precursor protein was used as a transport vehicle to test the tolerance of the mitochondrial protein import system. The precursor was a fusion protein consisting of mouse dihydrofolate reductase linked to a yeast mitochondrial presequence; it contained a unique cysteine as its COOH-terminal residue. This COOH-terminal cysteine was covalently coupled to either a stilbene disulfonate derivative or, with the aid of a bifunctional cross-linker, to one of the free amino groups of horse heart cytochrome c. Coupling to horse heart cytochrome c generated a mixture of branched polypeptide chains since this cytochrome lacks a free alpha-amino group. Both adducts were imported and cleaved by isolated yeast mitochondria. The mitochondrial protein import machinery can thus transport more complex structures and even highly charged "membrane-impermeant" organic molecules. This suggests that transport occurs through a hydrophilic environment.


Subject(s)
Intracellular Membranes/metabolism , Mitochondria/metabolism , Protein Precursors/metabolism , Biological Transport , Cytochrome c Group/metabolism , Electron Transport Complex IV/metabolism , In Vitro Techniques , Kinetics , Protein Precursors/ultrastructure , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/ultrastructure , Saccharomyces cerevisiae , Stilbenes/metabolism , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/metabolism
6.
J Cell Biol ; 147(5): 959-68, 1999 Nov 29.
Article in English | MEDLINE | ID: mdl-10579717

ABSTRACT

Translocation of nuclear-encoded preproteins across the outer membrane of mitochondria is mediated by the multicomponent transmembrane TOM complex. We have isolated the TOM core complex of Neurospora crassa by removing the receptors Tom70 and Tom20 from the isolated TOM holo complex by treatment with the detergent dodecyl maltoside. It consists of Tom40, Tom22, and the small Tom components, Tom6 and Tom7. This core complex was also purified directly from mitochondria after solubilization with dodecyl maltoside. The TOM core complex has the characteristics of the general insertion pore; it contains high-conductance channels and binds preprotein in a targeting sequence-dependent manner. It forms a double ring structure that, in contrast to the holo complex, lacks the third density seen in the latter particles. Three-dimensional reconstruction by electron tomography exhibits two open pores traversing the complex with a diameter of approximately 2.1 nm and a height of approximately 7 nm. Tom40 is the key structural element of the TOM core complex.


Subject(s)
Carrier Proteins/chemistry , Intracellular Membranes/enzymology , Membrane Proteins/chemistry , Mitochondria/enzymology , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Carrier Proteins/ultrastructure , Fungal Proteins/chemistry , Fungal Proteins/isolation & purification , Fungal Proteins/metabolism , Fungal Proteins/ultrastructure , Intracellular Membranes/metabolism , Intracellular Membranes/physiology , Intracellular Membranes/ultrastructure , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/physiology , Ion Channels/ultrastructure , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mitochondria/metabolism , Mitochondria/physiology , Mitochondria/ultrastructure , Neurospora crassa/enzymology , Neurospora crassa/metabolism , Neurospora crassa/physiology , Neurospora crassa/ultrastructure , Protein Binding , Protein Precursors/metabolism , Protein Precursors/ultrastructure
7.
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
8.
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
9.
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
10.
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
11.
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
12.
J Mol Biol ; 230(1): 260-83, 1993 Mar 05.
Article in English | MEDLINE | ID: mdl-8450540

ABSTRACT

The structure of mucor pusillus pepsin (EC 3.4.23.6), the aspartic proteinase from Mucor pusillus, has been refined to a crystallographic R-factor of 16.2% at 2.0 A resolution. The positions of 2638 protein atoms, 221 solvent atoms and a sulphate ion have been determined with an estimated root-mean-square (r.m.s.) error of 0.15 to 0.20 A. In the final model, the r.m.s. deviation from ideality for bond distances is 0.022 A, and for angle distances it is 0.050 A. Comparison of the overall three-dimensional structure with other aspartic proteinases shows that mucor pusillus pepsin is as distant from the other fungal enzymes as it is from those of mammalian origin. Analysis of a rigid body shift of residues 190 to 302 shows that mucor pusillus pepsin displays one of the largest shifts relative to other aspartic proteinases (14.4 degrees relative to endothiapepsin) and that changes have occurred at the interface between the two rigid bodies to accommodate this large shift. A new sequence alignment has been obtained on the basis of the three-dimensional structure, enabling the positions of large insertions to be identified. Analysis of secondary structure shows the beta-sheet to be well conserved whereas alpha-helical elements are more variable. A new alpha-helix hN4 is formed by a six-residue insertion between positions 131 and 132. Most insertions occur in loop regions: -5 to 1 (five residues relative to porcine pepsin): 115 to 116 (six residues); 186 to 187 (four residues); 263 to 264 (seven residues); 278 to 279 (four residues); and 326 to 332 (six residues). The active site residues are highly conserved in mucor pusillus pepsin; r.m.s. difference with rhizopuspepsin is 0.37 A for 25 C alpha atom pairs. However, residue 303, which is generally conserved as an aspartate, is changed to an asparagine in mucor pusillus pepsin, possibly influencing pH optimum. Substantial changes have occurred in the substrate binding cleft in the region of S1 and S3 due to the insertion between 115 and 116 and the rearrangement of loop 9-13. Residue Asn219 necessitates a shift in position of substrate main-chain atoms to maintain hydrogen bonding pattern. Invariant residues Asp11 and Tyr14 have undergone a major change in conformation apparently due to localized changes in molecular structure. Both these residues have been implicated in zymogen stability and activation.


Subject(s)
Aspartic Acid Endopeptidases/ultrastructure , Fungal Proteins/ultrastructure , Mucor/enzymology , Pepsin A/ultrastructure , Amino Acid Sequence , Binding Sites , Crystallography , Enzyme Activation , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Protein Precursors/ultrastructure , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Solvents , X-Ray Diffraction
13.
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
14.
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
15.
J Mol Biol ; 299(3): 573-84, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10835268

ABSTRACT

An assembly intermediate of a small, non-enveloped RNA virus has been discovered that exhibits striking differences from the mature virion. Virus-like particles (VLPs) of Nudaurelia capensis omega virus (NomegaV), a T=4 icosahedral virus infecting Lepidoptera insects, were produced in insect cells using a baculovirus vector expressing the coat protein. A procapsid form was discovered when NomegaV VLPs were purified at neutral pH conditions. These VLPs were fragile and did not undergo the autoproteolytic maturation that occurs in the infectious virus. Electron cryo-microscopy (cryoEM) and image analysis showed that, compared with the native virion, the VLPs were 16% larger in diameter, more rounded, porous, and contained an additional internal domain. Upon lowering the pH to 5.0, the VLP capsids became structurally indistinguishable from the authentic virion and the subunits autoproteolyzed. The NomegaV protein subunit coordinates, which were previously determined crystallographically, were modelled into the 28 A resolution cryoEM map of the procapsid. The resulting pseudo-atomic model of the NomegaV procapsid demonstrated the large rearrangements in quaternary and tertiary structure needed for the maturation of the VLPs and presumably of the virus. Based on this model, we propose that electrostatically driven rearrangements of interior helical regions are responsible for the large conformational change. These results are surprising because large structural rearrangements have not been found in the maturation of any other small RNA viruses. However, similarities of this conformational change to the maturational processes of more complex DNA viruses (e.g. bacteriophages and herpesvirus) and to the swelling of simple plant viruses suggest that structural changes in icosahedral viruses, which are integral to their function, have similar strategies and perhaps mechanisms.


Subject(s)
Capsid/chemistry , Insect Viruses/chemistry , Insect Viruses/ultrastructure , RNA Viruses/chemistry , RNA Viruses/ultrastructure , Virus Assembly , Animals , Binding Sites , Capsid/genetics , Capsid/metabolism , Capsid/ultrastructure , Cell Line , Cryoelectron Microscopy , Dimerization , Hydrogen-Ion Concentration , Insect Viruses/genetics , Insect Viruses/metabolism , Models, Molecular , Molecular Weight , Protein Conformation , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Protein Processing, Post-Translational , RNA Viruses/genetics , RNA Viruses/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Rotation , Spodoptera , Static Electricity
16.
Eur J Cell Biol ; 78(8): 593-600, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10494866

ABSTRACT

Nuclear envelope precursor vesicles were affinity purified from a Xenopus egg extract by a chromatin binding method. Vesicles bound to chromatin at 4 degrees C were dissociated with a high salt buffer and further fractionated into nuclear envelope precursor vesicle fractions 1 (PV1) and 2 (PV2) by differential centrifugation. PV1 contained larger vesicles. When chromatin was incubated in a Xenopus egg cytosol fraction supplemented with PV1, vesicles bound to chromatin, fused with each other, formed a bilayered nuclear envelope, and assembled into spherical small nuclei. However, the thus assembled nuclei did not grow to the normal size. Nuclear pore complexes were not found on the thus assembled nuclei. On the other hand, PV2 contained smaller vesicles. PV2 vesicles bound to chromatin, fused little with each other in the Xenopus egg cytosol fraction, and no nuclei were assembled. When PV1 supplemented with PV2 was used for the nuclear assembly reaction, the assembled nuclei grew to the normal size. Nuclear pore complexes existed in the thus assembled nuclear envelopes. These results suggested that 1) two vesicle populations, PV1 and PV2, are necessary for the assembly of normal sized nuclei, 2) PV1 contains a chromatin targeting molecule(s) and membrane fusion machinery, 3) PV2 contains a chromatin targeting molecule(s) and a molecule(s) necessary for nuclear pore complex assembly, and 4) PV1 has the ability to assemble a nuclear membrane, and PV2 is necessary for the assembly of nuclear pore complexes and for nuclei to grow to the normal size. An in vitro nuclear assembly system constituted with affinity-purified vesicle fractions, PV1 and PV2, was established.


Subject(s)
Cell Nucleus/metabolism , Nuclear Envelope/metabolism , Protein Precursors/isolation & purification , Animals , Cell Nucleus/ultrastructure , Chromatin/metabolism , Electrophoresis, Polyacrylamide Gel , Microscopy, Electron , Microscopy, Fluorescence , Nuclear Envelope/ultrastructure , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Xenopus/embryology
17.
J Invest Dermatol ; 98(6): 881-9, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1593151

ABSTRACT

Trichohyalin, a protein of Mr between 190 and 220 kDa in different species, was first demonstrated in large granules of the inner root sheath and medulla of hair follicles and may provide a matrix for keratin filaments. We have purified trichohyalin in milligram quantities from a citric acid-insoluble fraction derived from pig tongue epithelium. Trichohyalin was extracted under conditions of low ionic strength from the citric acid-insoluble fraction, separated by gel-filtration chromatography in buffer containing 1 M NaBr, and concentrated by ion-exchange chromatography in buffer containing 4 M urea. The purified material, which is soluble in buffers containing 1 M NaBr, was considered to be trichohyalin because of its characteristic molecular weight and amino acid composition and its localization to hair follicle inner root sheath and medulla by indirect immunofluorescence using antibodies against the purified protein. Immunofluorescence showed that trichohyalin is a major protein of filiform papillae of the tongue. Unlike trichohyalin from other animals examined, the porcine protein is a doublet on SDS polyacrylamide gels of 195 and 210 kDa; both bands are recognized by different antibodies, their two-dimensional peptide maps are nearly identical, and they have nearly identical isoelectric points of about 6.6. Trichohyalin has a Stokes radius of 124 A on gel filtration and a Svedberg constant of 6, consistent with an extended structure. The protein probably associates reversibly in solution, and the native protein we have isolated may be dimeric, because crosslinking of the iodinated purified protein with disuccinimidyl suberate demonstrated the presence of a dimer, which could be dissociated in the presence of high concentrations of urea. Rotary shadowing electron microscopy of the native protein showed a filamentous structure averaging 85 nm in length with a single globular-appearing end-domain. The purification of native trichohyalin provides a basis for future functional studies.


Subject(s)
Protein Precursors/isolation & purification , Tongue/chemistry , Amino Acids/analysis , Animals , Cross-Linking Reagents , Epithelium/chemistry , Intermediate Filament Proteins , Isoelectric Focusing , Microscopy, Electron , Peptide Mapping , Protein Precursors/chemistry , Protein Precursors/ultrastructure , Swine
18.
Gene ; 84(2): 303-9, 1989 Dec 14.
Article in English | MEDLINE | ID: mdl-2515117

ABSTRACT

The methylotrophic yeast, Hansenula polymorpha, has been developed as a host system for the synthesis of heterologous proteins. The middle surface antigen of hepatitis B virus (preS2-HBsAg) has been synthesized under the control of a methanol-regulated promoter derived from the methanol oxidase-encoding gene. The synthesized preS2-HBsAg protein was found to be secreted outside the cell membrane into the periplasm and further excreted into the culture medium following permeabilization of the cell wall with beta-1,3-glucanase (beta Glu). Cell cultures treated with beta Glu were able to continuously synthesize and secrete 22-nm particles of preS2-HBsAg into the medium for several days. The overall yield of antigen from treated cultures was found to be over threefold greater than that of untreated controls. The observation that complex supramolecular structures, such as the 22-nm particles of preS2-HBsAg, can be secreted by H. polymorpha and released into the medium, suggests the potential for these yeasts to be an alternative secretory host.


Subject(s)
Hepatitis B Surface Antigens/biosynthesis , Pichia/genetics , Saccharomycetales/genetics , Cloning, Molecular , Cytoplasm/metabolism , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Glucan 1,3-beta-Glucosidase , Hepatitis B Surface Antigens/metabolism , Immunoblotting , Plasmids , Protein Precursors/biosynthesis , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Transformation, Genetic , beta-Glucosidase
19.
AIDS Res Hum Retroviruses ; 7(5): 475-83, 1991 May.
Article in English | MEDLINE | ID: mdl-1873082

ABSTRACT

The vaccinia virus expression system was used to determine the role of human immunodeficiency virus type 1 (HIV-1) protease in viral morphogenesis and maturation. The unprocessed p55 gag precursor polyprotein alone was assembled to form HIV-1 particles which budded from cells. The particles were spherical and immature, containing an electron-dense shell in the particle submembrane; there was no evidence of core formation. Expression of both gag and pol proteins from a recombinant containing the complete gag-pol coding sequences resulted in intracellular processing of gag-pol proteins and the production of mature particles with electron-dense cores characteristic of wild-type HIV virions. To ascertain the role of protein processing in particle maturation, the pol ORF in the gag-pol recombinant was truncated to limit expression of the pol gene to the protease domain. With this recombinant expressing p55 gag and protease, intracellular processing was observed. Some of the resultant particles were partially mature and contained processed gag protein subunits. In contrast, particle maturation was not observed when the HIV-1 protease and p55 gag were coexpressed from separate recombinants, despite evidence of intracellular gag processing. These findings suggest that HIV-1 protease must be an integral component of the full-length gag-pol precursor for optimal processing and virion maturation.


Subject(s)
Fusion Proteins, gag-pol/metabolism , HIV Protease/metabolism , HIV-1/growth & development , Virion/growth & development , Virus Replication , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/ultrastructure , Gene Products, gag/genetics , Gene Products, gag/metabolism , Gene Products, gag/ultrastructure , HIV Protease/genetics , HIV Protease/ultrastructure , HIV-1/enzymology , HIV-1/ultrastructure , HeLa Cells , Humans , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Precursors/ultrastructure , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Recombination, Genetic , Vaccinia/genetics , Virion/ultrastructure
20.
Microsc Res Tech ; 27(4): 284-93, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8186447

ABSTRACT

Mitochondrial protein targeting includes both intramitochondrial sorting of proteins encoded by the organellar genome and import and subsequent sorting of nuclear encoded precursor proteins. Only a few proteins are encoded by the mitochondrial genome and synthesized in the organellar matrix. These include predominantly inner membrane proteins that are perhaps co-translationally inserted into this membrane. Biochemical data suggest that insertion into the inner membrane may be confined to the inner boundary membrane. Ultrastructurally, however, a preferential association of ribosomes with either inner boundary or cristae membranes has not been established. The majority of the mitochondrial proteins are nuclear encoded and synthesized as precursors in the cytosol. Electron microscopic studies revealed that import of precursor proteins is generally confined to sites where both mitochondrial envelope membranes are closely apposed. In line with these observations, biochemical studies indicated that precursor proteins destined for the inner membrane or matrix have to interact with the energized inner membrane to allow complete passage of the precursor through the outer membrane. As a consequence, the mitochondrial envelope membranes have to be in close proximity at protein import sites. In isolated mitochondria distinct sites (designated as contact sites) exist where both envelope membranes are closely apposed and presumably stably associated. In situ, however, mitochondrial boundary membranes are in close proximity over large areas that cover almost the entire mitochondrial periphery. Consequently, the relative area of the mitochondrial surface, where both boundary membranes are in sufficient proximity for allowing protein translocation, is generally larger in situ compared to that in isolated organelles. Immunocytochemical localization studies showed a rather random distribution of components of the mitochondrial protein translocation machinery over the entire mitochondrial surface and not confined to contact sites. Based on these ultrastructural data and recent biochemical findings we propose that mitochondrial protein import sites are dynamic in nature and include relatively labile regions of close association of the boundary membranes. In vitro, however, mitochondrial protein import may preferentially take place at or near the presumably stable contact sites.


Subject(s)
Mitochondria/ultrastructure , Protein Precursors/ultrastructure , Biological Transport , Candida/ultrastructure , Membrane Proteins/ultrastructure , Neurospora crassa/ultrastructure , Ribosomes/ultrastructure , Saccharomyces cerevisiae/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL