Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34635581

ABSTRACT

The host cell serine protease TMPRSS2 is an attractive therapeutic target for COVID-19 drug discovery. This protease activates the Spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and of other coronaviruses and is essential for viral spread in the lung. Utilizing rational structure-based drug design (SBDD) coupled to substrate specificity screening of TMPRSS2, we have discovered covalent small-molecule ketobenzothiazole (kbt) TMPRSS2 inhibitors which are structurally distinct from and have significantly improved activity over the existing known inhibitors Camostat and Nafamostat. Lead compound MM3122 (4) has an IC50 (half-maximal inhibitory concentration) of 340 pM against recombinant full-length TMPRSS2 protein, an EC50 (half-maximal effective concentration) of 430 pM in blocking host cell entry into Calu-3 human lung epithelial cells of a newly developed VSV-SARS-CoV-2 chimeric virus, and an EC50 of 74 nM in inhibiting cytopathic effects induced by SARS-CoV-2 virus in Calu-3 cells. Further, MM3122 blocks Middle East respiratory syndrome coronavirus (MERS-CoV) cell entry with an EC50 of 870 pM. MM3122 has excellent metabolic stability, safety, and pharmacokinetics in mice, with a half-life of 8.6 h in plasma and 7.5 h in lung tissue, making it suitable for in vivo efficacy evaluation and a promising drug candidate for COVID-19 treatment.


Subject(s)
Benzothiazoles/pharmacology , COVID-19 Drug Treatment , Oligopeptides/pharmacology , SARS-CoV-2/drug effects , Serine Endopeptidases/genetics , Animals , Benzamidines/chemistry , Benzothiazoles/pharmacokinetics , COVID-19/genetics , COVID-19/virology , Cell Line , Drug Design , Epithelial Cells/drug effects , Epithelial Cells/virology , Esters/chemistry , Guanidines/chemistry , Humans , Lung/drug effects , Lung/virology , Mice , Middle East Respiratory Syndrome Coronavirus/drug effects , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Oligopeptides/pharmacokinetics , SARS-CoV-2/pathogenicity , Serine Endopeptidases/drug effects , Serine Endopeptidases/ultrastructure , Small Molecule Libraries/pharmacology , Substrate Specificity/drug effects , Virus Internalization/drug effects
2.
Biochem J ; 477(2): 407-429, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31899476

ABSTRACT

Human HtrA3 (high-temperature requirement protease A3) is a trimeric multitasking propapoptotic serine protease associated with critical cellular functions and pathogenicity. Implicated in diseases including cancer and pre-eclampsia, its role as a tumor suppressor and potential therapeutic target cannot be ignored. Therefore, elucidating its mode of activation and regulatory switch becomes indispensable towards modulating its functions with desired effects for disease intervention. Using computational, biochemical and biophysical tools, we delineated the role of all domains, their combinations and the critical phenylalanine residues in regulating HtrA3 activity, oligomerization and specificity. Our findings underline the crucial roles of the N-terminus as well as the PDZ domain in oligomerization and formation of a catalytically competent enzyme, thus providing new insights into its structure-function coordination. Our study also reports an intricate ligand-induced allosteric switch, which redefines the existing hypothesis of HtrA3 activation besides opening up avenues for modulating protease activity favorably through suitable effector molecules.


Subject(s)
Protein Conformation , Serine Endopeptidases/genetics , Serine Proteases/genetics , Structure-Activity Relationship , Allosteric Regulation/genetics , Amino Acid Sequence/genetics , Catalysis , Gene Expression Regulation, Enzymologic/genetics , Humans , PDZ Domains/genetics , Protein Multimerization/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Serine Proteases/chemistry , Serine Proteases/ultrastructure
3.
Biochem Biophys Res Commun ; 487(2): 356-361, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28414130

ABSTRACT

Enzymes belonging to the S9 family of prolyl oligopeptidases are of interest because of their pharmacological importance and have a non-catalytic ß-propeller domain. In this study, we found that the oxidation of Met203, which lies on surface of the ß-propeller domain, leads to change in the substrate specificity of eryngase, an enzyme from Pleurotus eryngii and a member of the S9 family of prolyl oligopeptidases. The activity of eryngase for L-Phe-p-nitroanilide was maintained following hydrogen peroxide treatment but was dramatically reduced for other p-nitroanilide substrates. MALDI-TOF MS analysis using tryptic peptides of eryngase indicated that the change in substrate specificity was triggered by oxidizing Met203 to methionine sulfoxide. In addition, mutations of Met203 to smaller residues provided specificities similar to those observed following oxidation of the wild-type enzyme. Substitution of Met203 with Phe significantly decreased activity, indicating that Met203 may be involved in substrate gating.


Subject(s)
Molecular Docking Simulation , Oxygen/chemistry , Pleurotus/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Binding Sites , Enzyme Activation , Models, Chemical , Oxidation-Reduction , Prolyl Oligopeptidases , Protein Binding , Protein Conformation , Protein Domains , Protein Structure, Quaternary , Serine Endopeptidases/classification , Structure-Activity Relationship , Substrate Specificity
4.
Biochem Biophys Res Commun ; 492(4): 643-651, 2017 10 28.
Article in English | MEDLINE | ID: mdl-28341122

ABSTRACT

America is still suffering with the outbreak of Zika virus (ZIKV) infection. Congenital ZIKV syndrome has already caused a public health emergency of international concern. However, there are still no vaccines to prevent or drugs to treat the infection caused by ZIKV. The ZIKV NS3 helicase (NS3h) protein is a promising target for drug discovery due to its essential role in viral genome replication. NS3h unwinds the viral RNA to enable the replication of the viral genome by the NS5 protein. NS3h contains two important binding sites: the NTPase binding site and the RNA binding site. Here, we used molecular dynamics (MD) simulations to study the molecular behavior of ZIKV NS3h in the presence and absence of ssRNA and the potential implications for NS3h activity and inhibition. Although there is conformational variability and poor electron densities of the RNA binding loop in various apo flaviviruses NS3h crystallographic structures, the MD trajectories of NS3h-ssRNA demonstrated that the RNA binding loop becomes more stable when NS3h is occupied by RNA. Our results suggest that the presence of RNA generates important interactions with the RNA binding loop, and these interactions stabilize the loop sufficiently that it remains in a closed conformation. This closed conformation likely keeps the ssRNA bound to the protein for a sufficient duration to enable the unwinding/replication activities of NS3h to occur. In addition, conformational changes of this RNA binding loop can change the nature and location of the optimal ligand binding site, according to ligand binding site prediction results. These are important findings to help guide the design and discovery of new inhibitors of NS3h as promising compounds to treat the ZIKV infection.


Subject(s)
Models, Chemical , Molecular Dynamics Simulation , RNA, Viral/chemistry , RNA, Viral/ultrastructure , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/ultrastructure , Zika Virus/enzymology , Binding Sites , Enzyme Activation , Nucleic Acid Conformation , Protein Binding , Protein Conformation , RNA Helicases/chemistry , RNA Helicases/ultrastructure , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure
5.
Arch Biochem Biophys ; 621: 6-23, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28396256

ABSTRACT

Human HtrA1-4 proteins belong to the HtrA family of evolutionarily conserved serine proteases and function as important modulators of many physiological processes, including maintenance of mitochondrial homeostasis, cell signaling and apoptosis. Disturbances in their action are linked to severe diseases, including oncogenesis and neurodegeneration. The HtrA1-4 proteins share structural and functional features of other members of the HtrA protein family, however there are several significant differences in structural architecture and mechanisms of action which makes each of them unique. Our goal is to present recent studies regarding human HtrAs. We focus on their physiological functions, structure and regulation, and describe current models of activation mechanisms. Knowledge of molecular basis of the human HtrAs' action is a subject of great interest; it is crucial for understanding their relevance in cellular physiology and pathogenesis as well as for using them as targets in future therapies of diseases such as neurodegenerative disorders and cancer.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Binding Sites , Enzyme Activation , Humans , PDZ Domains/physiology , Protein Binding , Protein Conformation , Serine Endopeptidases/ultrastructure , Structure-Activity Relationship
6.
Biochem Biophys Res Commun ; 461(4): 677-80, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25918021

ABSTRACT

The dengue virus NS2B-NS3 protease (NS2B-NS3p), an important antiviral target for drug development, has been reported to adopt an open or closed conformation in crystal structures with different NS2B C-terminus (NS2Bc) positioning. In solution, nevertheless, NS2B-NS3p forms a mixture of open, closed and maybe other intermediate conformations, which is difficult to characterize using conventional biophysical and biochemical techniques. In this study, we developed a new strategy to analyze these conformational changes using (19)F NMR spectroscopy. Low pH or bovine pancreatic trypsin inhibitor (BPTI) binding promote the conformation change from open to closed, showing the importance of charge forces in the interaction between NS2Bc and NS3p. The mutation H51A impairs the charge interaction and the pH dependence of the conformational changes. It stabilizes the open conformation, while the addition of BPTI still converts NS2B-NS3p from open to closed conformation.


Subject(s)
Fluorine/analysis , Fluorine/chemistry , Magnetic Resonance Spectroscopy/methods , Models, Chemical , Models, Molecular , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Computer Simulation , Protein Conformation , Static Electricity
7.
Biochemistry ; 53(41): 6452-62, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25232897

ABSTRACT

Self-assembling proteins represent potential scaffolds for the organization of enzymatic activities. The alkaline protease repeats-in-toxin (RTX) domain from Pseudomonas aeruginosa undergoes multiple structural transitions in the presence and absence of calcium, a native structural cofactor. In the absence of calcium, this domain is capable of spontaneous, ordered polymerization, producing amyloid-like fibrils and large two-dimensional protein sheets. This polymerization occurs under near-physiological conditions, is rapid, and can be controlled by regulating calcium in solution. Fusion of the RTX domain to a soluble protein results in the incorporation of engineered protein function into these macromolecular assemblies. Applications of this protein sequence in bacterial adherence and colonization and the generation of biomaterials are discussed.


Subject(s)
Amyloid/chemistry , Bacterial Proteins/chemistry , Calcium/chemistry , Metalloexopeptidases/chemistry , Models, Molecular , Pseudomonas aeruginosa/enzymology , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/ultrastructure , Amyloid/genetics , Amyloid/metabolism , Amyloid/ultrastructure , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Calcium/metabolism , Circular Dichroism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/ultrastructure , Kinetics , Metalloexopeptidases/genetics , Metalloexopeptidases/metabolism , Metalloexopeptidases/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Polymerization , Protein Aggregation, Pathological , Protein Engineering , Protein Folding , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/ultrastructure , Repetitive Sequences, Amino Acid , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Serine Endopeptidases/ultrastructure
8.
Nature ; 453(7197): 885-90, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18496527

ABSTRACT

All organisms have to monitor the folding state of cellular proteins precisely. The heat-shock protein DegP is a protein quality control factor in the bacterial envelope that is involved in eliminating misfolded proteins and in the biogenesis of outer-membrane proteins. Here we describe the molecular mechanisms underlying the regulated protease and chaperone function of DegP from Escherichia coli. We show that binding of misfolded proteins transforms hexameric DegP into large, catalytically active 12-meric and 24-meric multimers. A structural analysis of these particles revealed that DegP represents a protein packaging device whose central compartment is adaptable to the size and concentration of substrate. Moreover, the inner cavity serves antagonistic functions. Whereas the encapsulation of folded protomers of outer-membrane proteins is protective and might allow safe transit through the periplasm, misfolded proteins are eliminated in the molecular reaction chamber. Oligomer reassembly and concomitant activation on substrate binding may also be critical in regulating other HtrA proteases implicated in protein-folding diseases.


Subject(s)
Escherichia coli/enzymology , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Periplasmic Proteins/chemistry , Periplasmic Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/ultrastructure , Cell Membrane/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Heat-Shock Proteins/ultrastructure , Models, Molecular , Molecular Chaperones/ultrastructure , Periplasmic Proteins/ultrastructure , Protein Folding , Protein Structure, Quaternary , Serine Endopeptidases/ultrastructure , Structure-Activity Relationship
9.
Proc Natl Acad Sci U S A ; 106(12): 4858-63, 2009 Mar 24.
Article in English | MEDLINE | ID: mdl-19255437

ABSTRACT

In the periplasm of Escherichia coli, DegP (also known as HtrA), which has both chaperone-like and proteolytic activities, prevents the accumulation of toxic misfolded and unfolded polypeptides. In solution, upon binding to denatured proteins, DegP forms large cage-like structures. Here, we show that DegP forms a range of bowl-shaped structures, independent of substrate proteins, each with a 4-, 5-, or 6-fold symmetry and all with a DegP trimer as the structural unit, on lipid membranes. These membrane-bound DegP assemblies have the capacity to recruit and process substrates in the bowl chamber, and they exhibit higher proteolytic and lower chaperone-like activities than DegP in solution. Our findings imply that DegP might regulate its dual roles during protein quality control, depending on its assembly state in the narrow bacterial envelope.


Subject(s)
Cell Membrane/enzymology , Escherichia coli/enzymology , Heat-Shock Proteins/chemistry , Periplasmic Proteins/chemistry , Serine Endopeptidases/chemistry , Cell Membrane/ultrastructure , Escherichia coli/cytology , Escherichia coli/ultrastructure , Heat-Shock Proteins/ultrastructure , Lipids/chemistry , Molecular Chaperones/metabolism , Periplasmic Proteins/ultrastructure , Protein Processing, Post-Translational , Protein Structure, Quaternary , Serine Endopeptidases/ultrastructure , Substrate Specificity
10.
J Biol Chem ; 285(42): 32336-42, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20688909

ABSTRACT

We present a body of ultrastructural, biochemical, and genetic evidence that demonstrates the oligomerization of virulence-associated autotransporter proteins EspC or EspP produced by deadly human pathogens enterohemorrhagic and enteropathogenic Escherichia coli into novel macroscopic rope-like structures (>1 cm long). The rope-like structures showed high aggregation and insolubility, stability to anionic detergents and high temperature, and binding to Congo Red and thioflavin T dyes. These are properties also exhibited by human amyloidogenic proteins. These macroscopic ropes were not observed in cultures of nonpathogenic Escherichia coli or isogenic espP or espC deletion mutants of enterohemorrhagic or enteropathogenic Escherichia coli but were produced by an Escherichia coli K-12 strain carrying a plasmid expressing espP. Purified recombinant EspP monomers were able to self-assemble into macroscopic ropes upon incubation, suggesting that no other protein was required for assembly. The ropes bound to and showed cytopathic effects on cultured epithelial cells, served as a substratum for bacterial adherence and biofilm formation, and protected bacteria from antimicrobial compounds. We hypothesize that these ropes play a biologically significant role in the survival and pathogenic scheme of these organisms.


Subject(s)
Bacterial Adhesion , Enterohemorrhagic Escherichia coli , Enteropathogenic Escherichia coli , Escherichia coli Proteins , Serine Endopeptidases , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Enterohemorrhagic Escherichia coli/chemistry , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Enteropathogenic Escherichia coli/chemistry , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/ultrastructure , HeLa Cells , Humans , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/ultrastructure
11.
Proc Natl Acad Sci U S A ; 105(33): 11939-44, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18697939

ABSTRACT

Cells use molecular chaperones and proteases to implement the essential quality control mechanism of proteins. The DegP (HtrA) protein, essential for the survival of Escherichia coli cells at elevated temperatures with homologues found in almost all organisms uniquely has both functions. Here we report a mechanism for DegP to activate both functions via formation of large cage-like 12- and 24-mers after binding to substrate proteins. Cryo-electron microscopic and biochemical studies revealed that both oligomers are consistently assembled by blocks of DegP trimers, via pairwise PDZ1-PDZ2 interactions between neighboring trimers. Such interactions simultaneously eliminate the inhibitory effects of the PDZ2 domain. Additionally, both DegP oligomers were also observed in extracts of E. coli cells, strongly implicating their physiological importance.


Subject(s)
Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Periplasmic Proteins/metabolism , Serine Endopeptidases/metabolism , Enzyme Activation , Escherichia coli/enzymology , Escherichia coli/genetics , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/ultrastructure , Microscopy, Electron , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/ultrastructure , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Periplasmic Proteins/ultrastructure , Protein Binding , Protein Folding , Protein Structure, Quaternary , Protein Structure, Tertiary , Serine Endopeptidases/chemistry , Serine Endopeptidases/classification , Serine Endopeptidases/genetics , Serine Endopeptidases/ultrastructure , Substrate Specificity
12.
Biochim Biophys Acta Proteins Proteom ; 1869(2): 140562, 2021 02.
Article in English | MEDLINE | ID: mdl-33221341

ABSTRACT

Affinity maturation of U33, a recombinant Fab inhibitor of uPA, was used to improve the affinity and the inhibitory effect compared to the parental Fab. Arginine scanning of the six CDR loops of U33 was done to identify initial binding determinants since uPA prefers arginine in its primary substrate binding pocket. Two CDR loops were selected to create an engineered affinity maturation library of U33 that was diversified around ArgL91 (CDR L3) and ArgH52 (CDR H2). Biopanning of the randomized U33 library under stringent conditions resulted in eight Fabs with improved binding properties. One of the most potent inhibitors, AB2, exhibited a 13-fold decrease in IC50 when compared to U33 largely due to a decrease in its off rate. To identify contributions of interfacial residues that might undergo structural rearrangement upon interface formation we used X-ray footprinting and mass spectrometry (XFMS). Four residues showed a pronounced decrease in solvent accessibility, and their clustering suggests that AB2 targets the active site and also engages residues in an adjacent pocket unique to human uPA. The 2.9 Å resolution crystal structure of AB2-bound to uPA shows a binding mode in which the CDR L1 loop inserts into the active site cleft and acts as a determinant of inhibition. The selectivity determinant of this binding mode is unlike previously identified inhibitory Fabs against uPA related serine proteases, MTSP-1, HGFA and FXIa. CDRs H2 and L3 loops aid in interface formation and provide critical salt-bridges to remodel loops surrounding the active site of uPA providing specificity and further evidence that antibodies can be potent and selective inhibitors of proteolytic enzymes.


Subject(s)
Recombinant Proteins/ultrastructure , Serine Proteases/chemistry , Serine Proteinase Inhibitors/chemistry , Urokinase-Type Plasminogen Activator/chemistry , Amino Acid Sequence/genetics , Humans , Quinuclidines/chemistry , Quinuclidines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Serine Proteases/genetics , Serine Proteinase Inhibitors/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics
13.
Nat Commun ; 11(1): 6063, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247098

ABSTRACT

Opportunistic pathogens such as Streptococcus pneumoniae secrete a giant metalloprotease virulence factor responsible for cleaving host IgA1, yet the molecular mechanism has remained unknown since their discovery nearly 30 years ago despite the potential for developing vaccines that target these enzymes to block infection. Here we show through a series of cryo-electron microscopy single particle reconstructions how the Streptococcus pneumoniae IgA1 protease facilitates IgA1 substrate recognition and how this can be inhibited. Specifically, the Streptococcus pneumoniae IgA1 protease subscribes to an active-site-gated mechanism where a domain undergoes a 10.0 Å movement to facilitate cleavage. Monoclonal antibody binding inhibits this conformational change, providing a direct means to block infection at the host interface. These structural studies explain decades of biological and biochemical studies and provides a general strategy to block Streptococcus pneumoniae IgA1 protease activity to potentially prevent infection.


Subject(s)
Protease Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Streptococcus pneumoniae/enzymology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/ultrastructure , Biocatalysis , Catalytic Domain , Cryoelectron Microscopy , Models, Molecular , Protein Binding , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure
14.
Biochim Biophys Acta Gen Subj ; 1864(7): 129581, 2020 07.
Article in English | MEDLINE | ID: mdl-32114025

ABSTRACT

BACKGROUND: In Gram-negative bacteria, type Va and Vc autotransporters are proteins that contain both a secreted virulence factor (the "passenger" domain) and a ß-barrel that aids its export. While it is known that the folding and insertion of the ß-barrel domain utilize the ß-barrel assembly machinery (BAM) complex, how the passenger domain is secreted and folded across the membrane remains to be determined. The hairpin model states that passenger domain secretion occurs independently through the fully-formed and membrane-inserted ß-barrel domain via a hairpin folding intermediate. In contrast, the BamA-assisted model states that the passenger domain is secreted through a hybrid of BamA, the essential subunit of the BAM complex, and the ß-barrel domain of the autotransporter. METHODS: To ascertain the models' plausibility, we have used molecular dynamics to simulate passenger domain secretion for two autotransporters, EspP and YadA. RESULTS: We observed that each protein's ß-barrel is unable to accommodate the secreting passenger domain in a hairpin configuration without major structural distortions. Additionally, the force required for secretion through EspP's ß-barrel is more than that through the BamA ß-barrel. CONCLUSIONS: Secretion of autotransporters most likely occurs through an incompletely formed ß-barrel domain of the autotransporter in conjunction with BamA. GENERAL SIGNIFICANCE: Secretion of virulence factors is a process used by practically all pathogenic Gram-negative bacteria. Understanding this process is a necessary step towards limiting their infectious capacity.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Protein Folding , Type V Secretion Systems/genetics , Bacterial Outer Membrane Proteins/ultrastructure , Biological Transport/genetics , Escherichia coli/genetics , Escherichia coli/ultrastructure , Escherichia coli Proteins/ultrastructure , Humans , Serine Endopeptidases/genetics , Serine Endopeptidases/ultrastructure
15.
Nat Struct Mol Biol ; 27(4): 323-332, 2020 04.
Article in English | MEDLINE | ID: mdl-32203490

ABSTRACT

Ribosome-associated quality control (RQC) represents a rescue pathway in eukaryotic cells that is triggered upon translational stalling. Collided ribosomes are recognized for subsequent dissociation followed by degradation of nascent peptides. However, endogenous RQC-inducing sequences and the mechanism underlying the ubiquitin-dependent ribosome dissociation remain poorly understood. Here, we identified SDD1 messenger RNA from Saccharomyces cerevisiae as an endogenous RQC substrate and reveal the mechanism of its mRNA-dependent and nascent peptide-dependent translational stalling. In vitro translation of SDD1 mRNA enabled the reconstitution of Hel2-dependent polyubiquitination of collided disomes and, preferentially, trisomes. The distinct trisome architecture, visualized using cryo-EM, provides the structural basis for the more-efficient recognition by Hel2 compared with that of disomes. Subsequently, the Slh1 helicase subunit of the RQC trigger (RQT) complex preferentially dissociates the first stalled polyubiquitinated ribosome in an ATP-dependent manner. Together, these findings provide fundamental mechanistic insights into RQC and its physiological role in maintaining cellular protein homeostasis.


Subject(s)
Cell Cycle Proteins/ultrastructure , Protein Biosynthesis , Ribosomes/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Serine Endopeptidases/ultrastructure , Ubiquitin-Protein Ligases/ultrastructure , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Peptides/chemistry , Peptides/genetics , RNA, Messenger/genetics , Ribosomes/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
16.
J Virol ; 82(17): 8838-48, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18562541

ABSTRACT

The hepatitis C virus (HCV) RNA replication complex (RC), which is composed of viral nonstructural (NS) proteins and host cellular proteins, replicates the viral RNA genome in association with intracellular membranes. Two viral NS proteins, NS3 and NS5A, are essential elements of the RC. Here, by using immunoprecipitation and fluorescence resonance energy transfer assays, we demonstrated that NS3 and NS5A interact with tubulin and actin. Furthermore, immunofluorescence microscopy and electron microscopy revealed that HCV RCs were aligned along microtubules and actin filaments in both HCV replicon cells and HCV-infected cells. In addition, the movement of RCs was inhibited when microtubules or actin filaments were depolymerized by colchicine and cytochalasin B, respectively. Based on our observations, we propose that microtubules and actin filaments provide the tracks for the movement of HCV RCs to other regions in the cell, and the molecular interactions between RCs and microtubules, or RCs and actin filaments, are mediated by NS3 and NS5A.


Subject(s)
Actin Cytoskeleton/metabolism , Microtubules/metabolism , Replicon , Viral Nonstructural Proteins/metabolism , Virus Replication , Actin Cytoskeleton/ultrastructure , Antibodies, Monoclonal/immunology , Carbocyanines/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Colchicine/pharmacology , Cytochalasin B/pharmacology , Fluorescein-5-isothiocyanate/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique, Direct , Fluorescent Dyes/metabolism , Hepacivirus/metabolism , Humans , Indoles/metabolism , Kidney/cytology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Microtubules/ultrastructure , RNA Helicases/metabolism , RNA Helicases/ultrastructure , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , Serine Endopeptidases/metabolism , Serine Endopeptidases/ultrastructure , Tubulin/metabolism , Tubulin Modulators/pharmacology , Viral Nonstructural Proteins/ultrastructure
17.
Virology ; 515: 74-80, 2018 02.
Article in English | MEDLINE | ID: mdl-29272748

ABSTRACT

Dengue virus (DENV) is an arbovirus, which replicates in the endoplasmic reticulum. Although replicative cycle takes place in the cytoplasm, some viral proteins such as NS5 and C are translocated to the nucleus during infection in mosquitoes and mammalian cells. To localized viral proteins in DENV-infected C6/36 cells, an immunofluorescence (IF) and immunoelectron microscopy (IEM) analysis were performed. Our results indicated that C, NS1, NS3 and NS5 proteins were found in the nucleus of DENV-infected C6/36 cells. Additionally, complex structures named strand-like structures (Ss) were observed in the nucleus of infected cells. Interestingly, the NS5 protein was located in these structures. Ss were absent in mock-infected cells, suggesting that DENV induces their formation in the nucleus of infected mosquito cells.


Subject(s)
Culicidae/virology , Dengue Virus/ultrastructure , Dengue/virology , Viral Nonstructural Proteins/ultrastructure , Animals , Cell Line , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Humans , Mice, Inbred BALB C , RNA Helicases/ultrastructure , Serine Endopeptidases/ultrastructure , Virus Replication
18.
BMC Struct Biol ; 7: 63, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17908326

ABSTRACT

BACKGROUND: A right-handed, calcium-dependent beta-roll structure found in secreted proteases and repeat-in-toxin proteins was used as a template for the design of minimal, soluble, monomeric polypeptides that would fold in the presence of Ca2+. Two polypeptides were synthesised to contain two and four metal-binding sites, respectively, and exploit stacked tryptophan pairs to stabilise the fold and report on the conformational state of the polypeptide. RESULTS: Initial analysis of the two polypeptides in the presence of calcium suggested the polypeptides were disordered. The addition of lanthanum to these peptides caused aggregation. Upon further study by right angle light scattering and electron microscopy, the aggregates were identified as ordered protein filaments that required lanthanum to polymerize. These filaments could be disassembled by the addition of a chelating agent. A simple head-to-tail model is proposed for filament formation that explains the metal ion-dependency. The model is supported by the capping of one of the polypeptides with biotin, which disrupts filament formation and provides the ability to control the average length of the filaments. CONCLUSION: Metal ion-dependent, reversible protein filament formation is demonstrated for two designed polypeptides. The polypeptides form filaments that are approximately 3 nm in diameter and several hundred nm in length. They are not amyloid-like in nature as demonstrated by their behaviour in the presence of congo red and thioflavin T. A capping strategy allows for the control of filament length and for potential applications including the "decoration" of a protein filament with various functional moieties.


Subject(s)
Lanthanum/pharmacology , Peptides/chemistry , Amino Acid Sequence , Amyloid/chemistry , Circular Dichroism , Light , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Polymers/chemistry , Protein Structure, Quaternary/drug effects , Protein Structure, Secondary/drug effects , Protein Structure, Tertiary , Pseudomonas aeruginosa/enzymology , Scattering, Radiation , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure
19.
J Comp Neurol ; 482(3): 294-308, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15690491

ABSTRACT

Reelin is a glycoprotein that plays a critical role in brain development, including proper cortical lamination. In adult animals, reelin continues to be expressed in different neuronal populations in many brain regions. We performed labeling for reelin immunoreactivity (-i) in post-mortem cerebral cortex from five adults and two fetuses with three different antibodies. The tissue was then processed for light and electron microscopy. In cell bodies, reelin-i was found in pyramidal and nonpyramidal neurons on the outer nuclear membrane, rough endoplasmic reticulum (rER), and ribosomes. In dendrites, labeling was found in the rER and ribosomes and was diffusely distributed in spines. In the neuropil, diffuse labeling was seen in small axon terminals and unmyelinated axons, and the postsynaptic density (PSD) frequently had discrete labeling. Reelin-i was also found in glial somata and in small astrocytic processes. With rare exceptions, reelin-i in the adult was conspicuously absent from both the extracellular matrix (ECM) and the subcellular organelles, where secreted proteins are modified and taken back into the cell. Labeling in fetal cortex was similar to that in the adult except for prominent labeling in the ECM. The presence of reelin in adult spines, PSD, and terminals suggests that in the adult human reelin has a role in synaptic remodeling, which is consistent with the evidence for its role in long-term potentiation in the adult brain.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/ultrastructure , Cerebral Cortex/ultrastructure , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/ultrastructure , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Serine Endopeptidases/metabolism , Serine Endopeptidases/ultrastructure , Adult , Aged , Aged, 80 and over , Cerebral Cortex/metabolism , Female , Fetus , Humans , Immunohistochemistry , Male , Middle Aged , Neuroglia/metabolism , Neurons/metabolism , Postmortem Changes , Reelin Protein , Reference Values , Tissue Distribution
20.
J Mol Biol ; 252(1): 20-4, 1995 Sep 08.
Article in English | MEDLINE | ID: mdl-7666429

ABSTRACT

The crystal structure of a cleaved form of porcine zymogen E has been solved by molecular replacement using the bovine procarboxypeptidase A-S6 subunit III structure as search model. Crystallographic refinement using simulated annealing and energy minimization techniques resulted in a final R-factor of 0.189 for all data between 8 and 2.3 A resolution. The zymogen E three-dimensional model is very close to that of bovine subunit III and represents the second member of the zymogen E family for which the crystal structure is known. The two structures indicate that, in contrast to trypsinogen and chymotrypsinogen, zymogens of this family are highly organized molecules. The amino acid sequence of zymogen E has only been determined for the first 40 residues. Based on the electron density map, we have introduced six sequence changes relative to subunit III. Out of the 11 residues in the activation peptide, only the first six present well matching electron density; they are connected to the rest of the zymogen by an unexpected Cys1-Cys122 disulphide bridge (according to the bovine chymotrypsinogen A numbering system). Amino acid sequencing of protein solutions both from dissolved crystals and from the initial stock clearly indicated that the Val17-Asn18 bond had been cleaved during the crystallization process. This result adds weight to the assumption that the autolysis of the bovine zymogen E gives rise to subunit III and that this maybe a regulatory mechanism for protease E activity.


Subject(s)
Enzyme Precursors/ultrastructure , Serine Endopeptidases/ultrastructure , Amino Acid Sequence , Animals , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid , Swine
SELECTION OF CITATIONS
SEARCH DETAIL