Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Cell Mol Life Sci ; 80(8): 214, 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37466729

ABSTRACT

Mutations in the photoreceptor-specific tetraspanin gene peripherin-2 (PRPH2) lead to widely varying forms of retinal degeneration ranging from retinitis pigmentosa to macular dystrophy. Both inter- and intra-familial phenotypic heterogeneity has led to much interest in uncovering the complex pathogenic mechanisms of PRPH2-associated disease. Majority of disease-causing mutations in PRPH2 reside in the second intradiscal loop, wherein seven cysteines control protein folding and oligomerization. Here, we utilize knockin models to evaluate the role of three D2 loop cysteine mutants (Y141C, C213Y and C150S), alone or in combination. We elucidated how these mutations affect PRPH2 properties, including oligomerization and subcellular localization, and contribute to disease processes. Results from our structural, functional and molecular studies revealed that, in contrast to our understanding from prior investigations, rods are highly affected by PRPH2 mutations interfering with oligomerization and not merely by the haploinsufficiency associated with these mutations. On the other hand, cones are less affected by the toxicity of the mutant protein and significantly reduced protein levels, suggesting that knockdown therapeutic strategies may sustain cone functionality for a longer period. This observation provides useful data to guide and simplify the current development of effective therapeutic approaches for PRPH2-associated diseases that combine knockdown with high levels of gene supplementation needed to generate prolonged rod improvement.


Subject(s)
Macular Degeneration , Retinal Degeneration , Retinitis Pigmentosa , Humans , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinitis Pigmentosa/metabolism , Macular Degeneration/pathology , Tetraspanins/metabolism , Mutation/genetics
3.
mBio ; 12(6): e0283321, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34724811

ABSTRACT

Shigella spp. are highly adapted pathogens that cause bacillary dysentery in human and nonhuman primates. An unusual feature of Shigella pathogenesis is that this organism invades the colonic epithelia from the basolateral pole. Therefore, it has evolved the ability to disrupt the intestinal epithelial barrier to reach the basolateral surface. We have shown previously that the secreted serine protease A (SepA), which belongs to the family of serine protease autotransporters of Enterobacteriaceae, is responsible for the initial destabilization of the intestinal epithelial barrier that facilitates Shigella invasion. However, the mechanisms used by SepA to regulate this process remain unknown. To investigate the protein targets cleaved by SepA in the intestinal epithelium, we incubated a sample of homogenized human colon with purified SepA or with a catalytically inactive mutant of this protease. We discovered that SepA targets an array of 18 different proteins, including alpha-1 antitrypsin (AAT), a major circulating serine proteinase inhibitor in humans. In contrast to other serine proteases, SepA cleaved AAT without forming an inhibiting complex, which resulted in the generation of a neutrophil chemoattractant. We demonstrated that the products of the AAT-SepA reaction induce a mild but significant increase in neutrophil transepithelial migration in vitro. Moreover, the presence of AAT during Shigella infection stimulated neutrophil migration and dramatically enhanced the number of bacteria invading the intestinal epithelium in a SepA-dependent manner. We conclude that by cleaving AAT, SepA releases a chemoattractant that promotes neutrophil migration, which in turn disrupts the intestinal epithelial barrier to enable Shigella invasion. IMPORTANCEShigella is the second leading cause of diarrheal death globally. In this study, we identified the host protein targets of SepA, Shigella's major protein secreted in culture. We demonstrated that by cleaving AAT, a serine protease inhibitor important to protect surrounding tissue at inflammatory sites, SepA releases a neutrophil chemoattractant that enhances Shigella invasion. Moreover, SepA degraded AAT without becoming inhibited by the cleaved product, and SepA catalytic activity was enhanced at higher concentrations of AAT. Activation of SepA by an excess of AAT may be physiologically relevant at the early stages of Shigella infection, when the amount of synthesized SepA is very low compared to the concentration of AAT in the intestinal lumen. This observation may also help to explain the adeptness of Shigella infectivity at low dose, despite the requirement of reaching the basolateral side to invade and colonize the colonic epithelium.


Subject(s)
Bacterial Proteins/metabolism , Chemotactic Factors/metabolism , Dysentery, Bacillary/metabolism , Epithelial Cells/microbiology , Neutrophils/cytology , Shigella/enzymology , alpha 1-Antitrypsin/metabolism , Bacterial Proteins/genetics , Cell Movement , Chemotactic Factors/genetics , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Epithelial Cells/metabolism , Humans , Intestines/cytology , Intestines/metabolism , Intestines/microbiology , Neutrophils/metabolism , Shigella/classification , Shigella/genetics , alpha 1-Antitrypsin/genetics
4.
ACS Chem Biol ; 16(9): 1757-1769, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34406751

ABSTRACT

Cysteine-rich knob domains found in the ultralong complementarity determining regions of a subset of bovine antibodies are capable of functioning autonomously as 3-6 kDa peptides. While they can be expressed recombinantly in cellular systems, in this paper we show that knob domains are also readily amenable to a chemical synthesis, with a co-crystal structure of a chemically synthesized knob domain in complex with an antigen showing structural equivalence to the biological product. For drug discovery, following the immunization of cattle, knob domain peptides can be synthesized directly from antibody sequence data, combining the power and diversity of the bovine immune repertoire with the ability to rapidly incorporate nonbiological modifications. We demonstrate that, through rational design with non-natural amino acids, a paratope diversity can be massively expanded, in this case improving the efficacy of an allosteric peptide. As a potential route to further improve stability, we also performed head-to-tail cyclizations, exploiting the proximity of the N and C termini to synthesize functional, fully cyclic antibody fragments. Lastly, we highlight the stability of knob domains in plasma and, through pharmacokinetic studies, use palmitoylation as a route to extend the plasma half-life of knob domains in vivo. This study presents an antibody-derived medicinal chemistry platform, with protocols for solid-phase synthesis of knob domains, together with the characterization of their molecular structures, in vitro pharmacology, and pharmacokinetics.


Subject(s)
Complementarity Determining Regions/chemistry , Immunoglobulin Fragments/chemistry , Peptides, Cyclic/chemical synthesis , Amino Acid Sequence , Animals , Cattle , Immunoglobulin Fragments/blood , Immunoglobulin Fragments/pharmacology , Male , Models, Molecular , Peptides, Cyclic/blood , Peptides, Cyclic/pharmacokinetics , Protein Binding , Protein Domains , Protein Folding , Rats, Sprague-Dawley , Solid-Phase Synthesis Techniques , Tandem Mass Spectrometry , Thermodynamics
5.
Elife ; 102021 02 11.
Article in English | MEDLINE | ID: mdl-33570492

ABSTRACT

Bovines have evolved a subset of antibodies with ultra-long heavy chain complementarity determining regions that harbour cysteine-rich knob domains. To produce high-affinity peptides, we previously isolated autonomous 3-6 kDa knob domains from bovine antibodies. Here, we show that binding of four knob domain peptides elicits a range of effects on the clinically validated drug target complement C5. Allosteric mechanisms predominated, with one peptide selectively inhibiting C5 cleavage by the alternative pathway C5 convertase, revealing a targetable mechanistic difference between the classical and alternative pathway C5 convertases. Taking a hybrid biophysical approach, we present C5-knob domain co-crystal structures and, by solution methods, observed allosteric effects propagating >50 Å from the binding sites. This study expands the therapeutic scope of C5, presents new inhibitors, and introduces knob domains as new, low molecular weight antibody fragments, with therapeutic potential.


Antibodies are proteins produced by the immune system that can selectively bind to other molecules and modify their behaviour. Cows are highly equipped at fighting-off disease-causing microbes due to the unique shape of some of their antibodies. Unlike other jawed vertebrates, cows' antibodies contain an ultra-long loop region that contains a 'knob domain' which sticks out from the rest of the antibody. Recent research has shown that when detached, the knob domain behaves like an antibody fragment, and can independently bind to a range of different proteins. Antibody fragments are commonly developed in the laboratory to target proteins associated with certain diseases, such as arthritis and cancer. But it was unclear whether the knob domains from cows' antibodies could also have therapeutic potential. To investigate this, Macpherson et al. studied how knob domains attach to complement C5, a protein in the inflammatory pathway which is a drug target for various diseases, including severe COVID-19. The experiments identified various knob domains that bind to complement C5 and inhibits its activity by altering its structure or movement. Further tests studying the structure of these interactions, led to the discovery of a common mechanism by which inhibitors can modify the behaviour of this inflammatory protein. Complement C5 is involved in numerous molecular pathways in the immune system, which means many of the drugs developed to inhibit its activity can also leave patients vulnerable to infection. However, one of the knob domains identified by Macpherson et al. was found to reduce the activity of complement C5 in some pathways, whilst leaving other pathways intact. This could potentially reduce the risk of bacterial infections which sometimes arise following treatment with these types of inhibitors. These findings highlight a new approach for developing drug inhibitors for complement C5. Furthermore, the ability of knob domains to bind to multiple sites of complement C5 suggests that this fragment could be used to target proteins associated with other diseases.


Subject(s)
Allosteric Regulation/drug effects , Complement C5/antagonists & inhibitors , Drug Discovery , Peptides/chemistry , Peptides/pharmacology , Animals , Cattle , Complement C5/chemistry , Complement C5/metabolism , Molecular Docking Simulation , Protein Conformation/drug effects
6.
Cancer Gene Ther ; 28(1-2): 18-26, 2021 02.
Article in English | MEDLINE | ID: mdl-32595215

ABSTRACT

OPCML is a highly conserved glycosyl phosphatidylinositol (GPI)-anchored protein belonging to the IgLON family of cell adhesion molecules. OPCML functions as a tumor suppressor and is silenced in over 80% of ovarian cancers by loss of heterozygosity and by epigenetic mechanisms. OPCML inactivation is also observed in many other cancers suggesting a conservation of tumor suppressor function. Although epigenetic silencing and subsequent loss of OPCML expression correlate with poor progression-free and overall patient survival, its mechanism of action is only starting to be fully elucidated. Recent discoveries have demonstrated that OPCML exerts its tumor suppressor effect by inhibiting several cancer hallmark phenotypes in vitro and abrogating tumorigenesis in vivo, by downregulating/inactivating a specific spectrum of Receptor Tyrosine Kinases (RTKs), including EphA2, FGFR1, FGFR3, HER2, HER4, and AXL. This modulation of RTKs can also sensitize ovarian and breast cancers to lapatinib, erlotinib, and anti-AXL therapies. Furthermore, OPCML has also been shown to function in synergy with the tumor suppressor phosphatase PTPRG to inactivate pro-metastatic RTKs such as AXL. Recently, the identification of inactivating point mutations and the elucidation of the crystal structure of OPCML have provided valuable insights into its structure-function relationships, giving rise to its potential as an anti-cancer therapeutic.


Subject(s)
Cell Adhesion Molecules/drug effects , Glycosylphosphatidylinositols/therapeutic use , Neoplasms/drug therapy , GPI-Linked Proteins/drug effects , Glycosylphosphatidylinositols/pharmacology , Humans
8.
Nat Commun ; 10(1): 5247, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748512

ABSTRACT

Individuals with narcolepsy suffer from abnormal sleep patterns due to loss of neurons that uniquely supply hypocretin (HCRT). Previous studies found associations of narcolepsy with the human leukocyte antigen (HLA)-DQ6 allele and T-cell receptor α (TRA) J24 gene segment and also suggested that in vitro-stimulated T cells can target HCRT. Here, we present evidence of in vivo expansion of DQ6-HCRT tetramer+/TRAJ24+/CD4+ T cells in DQ6+ individuals with and without narcolepsy. We identify related TRAJ24+ TCRαß clonotypes encoded by identical α/ß gene regions from two patients and two controls. TRAJ24-G allele+ clonotypes only expand in the two patients, whereas a TRAJ24-C allele+ clonotype expands in a control. A representative tetramer+/G-allele+ TCR shows signaling reactivity to the epitope HCRT87-97. Clonally expanded G-allele+ T cells exhibit an unconventional effector phenotype. Our analysis of in vivo expansion of HCRT-reactive TRAJ24+ cells opens an avenue for further investigation of the autoimmune contribution to narcolepsy development.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Narcolepsy/immunology , Orexins/immunology , Animals , Autoimmunity/genetics , Case-Control Studies , Cell Proliferation , Crystallography, X-Ray , Drosophila , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Humans , Immunoglobulin Joining Region/genetics , Narcolepsy/genetics , Peripheral Tolerance , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
9.
Nat Commun ; 10(1): 3134, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31316070

ABSTRACT

OPCML, a tumor suppressor gene, is frequently silenced epigenetically in ovarian and other cancers. Here we report, by analysis of databases of tumor sequences, the observation of OPCML somatic missense mutations from various tumor types and the impact of these mutations on OPCML function, by solving the X-ray crystal structure of this glycoprotein to 2.65 Å resolution. OPCML consists of an extended arrangement of three immunoglobulin-like domains and homodimerizes via a network of contacts between membrane-distal domains. We report the generation of a panel of OPCML variants with representative clinical mutations and demonstrate clear phenotypic effects in vitro and in vivo including changes to anchorage-independent growth, interaction with activated cognate receptor tyrosine kinases, cellular migration, invasion in vitro and tumor growth in vivo. Our results suggest that clinically occurring somatic missense mutations in OPCML have the potential to contribute to tumorigenesis in a variety of cancers.


Subject(s)
Cell Adhesion Molecules/genetics , Epigenesis, Genetic , Ovarian Neoplasms/genetics , Cell Adhesion Molecules/chemistry , Cell Transformation, Neoplastic , Crystallography, X-Ray , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Glycosylation , Humans , Mutation, Missense , Neoplasm Invasiveness , Protein Aggregation, Pathological/genetics , Protein Structure, Tertiary
10.
Gut Microbes ; 8(6): 544-560, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28598765

ABSTRACT

Shigella is unique among enteric pathogens, as it invades colonic epithelia through the basolateral pole. Therefore, it has evolved the ability to breach the intestinal epithelial barrier to deploy an arsenal of effector proteins, which permits bacterial invasion and leads to a severe inflammatory response. However, the mechanisms used by Shigella to regulate epithelial barrier permeability remain unknown. To address this question, we used both an intestinal polarized model and a human ex-vivo model to further characterize the early events of host-bacteria interactions. Our results showed that secreted Serine Protease A (SepA), which belongs to the serine protease autotransporter of Enterobacteriaceae family, is responsible for critically disrupting the intestinal epithelial barrier. Such disruption facilitates bacterial transit to the basolateral pole of the epithelium, ultimately fostering the hallmarks of the disease pathology. SepA was found to cause a decrease in active LIM Kinase 1 (LIMK1) levels, a negative inhibitor of actin-remodeling proteins, namely cofilin. Correspondingly, we observed increased activation of cofilin, a major actin-polymerization factor known to control opening of tight junctions at the epithelial barrier. Furthermore, we resolved the crystal structure of SepA to elucidate its role on actin-dynamics and barrier disruption. The serine protease activity of SepA was found to be required for the regulatory effects on LIMK1 and cofilin, resulting in the disruption of the epithelial barrier during infection. Altogether, we demonstrate that SepA is indispensable for barrier disruption, ultimately facilitating Shigella transit to the basolateral pole where it effectively invades the epithelium.


Subject(s)
Actin Depolymerizing Factors/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Shigella flexneri/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line, Tumor , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Models, Biological , Mutation , Neutrophil Infiltration/immunology , Permeability , Phosphorylation , Protein Structure, Secondary , Shigella flexneri/genetics , Shigella flexneri/immunology , Structure-Activity Relationship , Tight Junctions/immunology , Tight Junctions/metabolism , Tight Junctions/microbiology
11.
Transgenic Res ; 24(4): 665-91, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26048593

ABSTRACT

Recent reports of a new generation of ubiquitous transgenic chimaera markers prompted us to consider the criteria used to evaluate new chimaera markers and develop more objective assessment methods. To investigate this experimentally we used several series of fetal and adult chimaeras, carrying an older, multi-copy transgenic marker. We used two additional independent markers and objective, quantitative criteria for cell selection and cell mixing to investigate quantitative and spatial aspects of developmental neutrality. We also suggest how the quantitative analysis we used could be simplified for future use with other markers. As a result, we recommend a five-step procedure for investigators to evaluate new chimaera markers based partly on criteria proposed previously but with a greater emphasis on examining the developmental neutrality of prospective new markers. These five steps comprise (1) review of published information, (2) evaluation of marker detection, (3) genetic crosses to check for effects on viability and growth, (4) comparisons of chimaeras with and without the marker and (5) analysis of chimaeras with both cell populations labelled. Finally, we review a number of different chimaera markers and evaluate them using the extended set of criteria. These comparisons indicate that, although the new generation of ubiquitous fluorescent markers are the best of those currently available and fulfil most of the criteria required of a chimaera marker, further work is required to determine whether they are developmentally neutral.


Subject(s)
Biomarkers/metabolism , Chimera/genetics , Fetal Development/genetics , Gene Expression Regulation, Developmental , Genetic Markers , Retinal Degeneration/genetics , Transgenes/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Organ Specificity
12.
Proc Natl Acad Sci U S A ; 110(47): E4427-36, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24191035

ABSTRACT

Intrinsically disordered proteins play crucial roles in regulatory processes and often function as protein interaction hubs. Here, we present a detailed characterization of a full-length disordered hub protein region involved in multiple dynamic complexes. We performed NMR, CD, and fluorescence binding studies on the nonphosphorylated and highly PKA-phosphorylated human cystic fibrosis transmembrane conductance regulator (CFTR) regulatory region, a ∼200-residue disordered segment involved in phosphorylation-dependent regulation of channel trafficking and gating. Our data provide evidence for dynamic, phosphorylation-dependent, multisite interactions of various segments of the regulatory region for its intra- and intermolecular partners, including the CFTR nucleotide binding domains 1 and 2, a 42-residue peptide from the C terminus of CFTR, the SLC26A3 sulphate transporter and antisigma factor antagonist (STAS) domain, and 14-3-3ß. Because of its large number of binding partners, multivalent binding of individually weak sites facilitates rapid exchange between free and bound states to allow the regulatory region to engage with different partners and generate a graded or rheostat-like response to phosphorylation. Our results enrich the understanding of how disordered binding segments interact with multiple targets. We present structural models consistent with our data that illustrate this dynamic aspect of phospho-regulation of CFTR by the disordered regulatory region.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Models, Molecular , Protein Conformation , Protein Interaction Maps/physiology , Regulatory Sequences, Nucleic Acid/physiology , 14-3-3 Proteins/metabolism , Biophysics , Chloride-Bicarbonate Antiporters/metabolism , Circular Dichroism , Fluorescence , Humans , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Binding , Protein Folding , Protein Interaction Maps/genetics , Regulatory Sequences, Nucleic Acid/genetics , Sulfate Transporters
13.
Proc Natl Acad Sci U S A ; 110(49): 19890-5, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248368

ABSTRACT

Intracellular aminopeptidases endoplasmic reticulum aminopeptidases 1 and 2 (ERAP1 and ERAP2), and as well as insulin-regulated aminopeptidase (IRAP) process antigenic epitope precursors for loading onto MHC class I molecules and regulate the adaptive immune response. Their activity greatly affects the antigenic peptide repertoire presented to cytotoxic T lymphocytes and as a result can regulate cytotoxic cellular responses contributing to autoimmunity or immune evasion by viruses and cancer cells. Therefore, pharmacological regulation of their activity is a promising avenue for modulating the adaptive immune response with possible applications in controlling autoimmunity, in boosting immune responses to pathogens, and in cancer immunotherapy. In this study we exploited recent structural and biochemical analysis of ERAP1 and ERAP2 to design and develop phosphinic pseudopeptide transition state analogs that can inhibit this family of enzymes with nM affinity. X-ray crystallographic analysis of one such inhibitor in complex with ERAP2 validated our design, revealing a canonical mode of binding in the active site of the enzyme, and highlighted the importance of the S2' pocket for achieving inhibitor potency. Antigen processing and presentation assays in HeLa and murine colon carcinoma (CT26) cells showed that these inhibitors induce increased cell-surface antigen presentation of transfected and endogenous antigens and enhance cytotoxic T-cell responses, indicating that these enzymes primarily destroy epitopes in those systems. This class of inhibitors constitutes a promising tool for controlling the cellular adaptive immune response in humans by modulating the antigen processing and presentation pathway.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antigen Presentation/immunology , Models, Molecular , T-Lymphocytes, Cytotoxic/immunology , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Animals , Antigen Presentation/drug effects , Binding Sites/immunology , Cell Line, Tumor , Crystallography, X-Ray , Cystinyl Aminopeptidase/metabolism , HeLa Cells , Humans , Mice , Minor Histocompatibility Antigens , Molecular Structure , Phosphinic Acids , Protein Engineering , T-Lymphocytes, Cytotoxic/drug effects
14.
J Virol ; 87(10): 5318-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23487472

ABSTRACT

We report the solution structures of the VPg proteins from feline calicivirus (FCV) and murine norovirus (MNV), which have been determined by nuclear magnetic resonance spectroscopy. In both cases, the core of the protein adopts a compact helical structure flanked by flexible N and C termini. Remarkably, while the core of FCV VPg contains a well-defined three-helix bundle, the MNV VPg core has just the first two of these secondary structure elements. In both cases, the VPg cores are stabilized by networks of hydrophobic and salt bridge interactions. The Tyr residue in VPg that is nucleotidylated by the viral NS7 polymerase (Y24 in FCV, Y26 in MNV) occurs in a conserved position within the first helix of the core. Intriguingly, given its structure, VPg would appear to be unable to bind to the viral polymerase so as to place this Tyr in the active site without a major conformation change to VPg or the polymerase. However, mutations that destabilized the VPg core either had no effect on or reduced both the ability of the protein to be nucleotidylated and virus infectivity and did not reveal a clear structure-activity relationship. The precise role of the calicivirus VPg core in virus replication remains to be determined, but knowledge of its structure will facilitate future investigations.


Subject(s)
Calicivirus, Feline/chemistry , Norovirus/chemistry , Viral Proteins/chemistry , Animals , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Conformation
15.
Biochemistry ; 51(1): 286-95, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22106953

ABSTRACT

Endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 cooperate to trim a vast variety of antigenic peptide precursors to generate mature epitopes for binding to major histocompatibility class I molecules. We report here the first structure of ERAP2 determined at 3.08 Å by X-ray crystallography. On the basis of residual electron density, a lysine residue has been modeled in the active site of the enzyme; thus, the structure corresponds to an enzyme-product complex. The overall domain organization is highly similar to that of the recently determined structure of ERAP1 in its closed conformation. A large internal cavity adjacent to the catalytic site can accommodate large peptide substrates. The ERAP2 structure provides a structural explanation for the different peptide N-terminal specificities between ERAP1 and ERAP2 and suggests that such differences extend throughout the whole peptide sequence. A noncrystallographic dimer observed may constitute a model for a proposed ERAP1-ERAP2 heterodimer. Overall, the structure helps explain how two homologous aminopeptidases cooperate to process a large variety of sequences, a key property of their biological role.


Subject(s)
Aminopeptidases/chemistry , Antigen Presentation/immunology , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/immunology , Aminopeptidases/metabolism , Aminopeptidases/physiology , Crystallography, X-Ray , Glycosylation , HLA Antigens/chemistry , HLA Antigens/physiology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/physiology , Humans , Minor Histocompatibility Antigens , Models, Molecular , Peptides/chemistry , Peptides/physiology , Protein Conformation , Protein Precursors/chemistry , Protein Precursors/physiology , Protein Structure, Tertiary/physiology
16.
J Struct Biol ; 167(3): 242-51, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524678

ABSTRACT

We describe biochemical and structural studies of the isolated cystic fibrosis transmembrane conductance regulator (CFTR) protein. Using electron cryomicroscopy, low resolution three-dimensional structures have been obtained for the non-phosphorylated protein in the absence of nucleotide and for the phosphorylated protein with ATP. In the latter state, the cytosolic nucleotide-binding domains move closer together, forming a more compact packing arrangement. Associated with this is a reorganization within the cylindrical transmembrane domains, consistent with a shift from an inward-facing to outward-facing configuration. A region of density in the non-phosphorylated protein that extends from the bottom of the cytosolic regions up to the transmembrane domains is hypothesised to represent the unique regulatory region of CFTR. These data offer insights into the architecture of this ATP-binding cassette protein, and shed light on the global motions associated with nucleotide binding and priming of the chloride channel via phosphorylation of the regulatory region.


Subject(s)
Adenosine Triphosphate/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Protein Processing, Post-Translational , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Phosphorylation , Protein Conformation , Protein Multimerization
17.
EMBO J ; 26(17): 3936-44, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17690690

ABSTRACT

The ErbB3-binding protein 1 (Ebp1) is an important regulator of transcription, affecting eukaryotic cell growth, proliferation, differentiation and survival. Ebp1 can also affect translation and cooperates with the polypyrimidine tract-binding protein (PTB) to stimulate the activity of the internal ribosome entry site (IRES) of foot-and-mouth disease virus (FMDV). We report here the crystal structure of murine Ebp1 (p48 isoform), providing the first glimpse of the architecture of this versatile regulator. The structure reveals a core domain that is homologous to methionine aminopeptidases, coupled to a C-terminal extension that contains important motifs for binding proteins and RNA. It sheds new light on the conformational differences between the p42 and p48 isoforms of Ebp1, the disposition of the key protein-interacting motif ((354)LKALL(358)) and the RNA-binding activity of Ebp1. We show that the primary RNA-binding site is formed by a Lys-rich motif in the C terminus and mediates the interaction with the FMDV IRES. We also demonstrate a specific functional requirement for Ebp1 in FMDV IRES-directed translation that is independent of a direct interaction with PTB.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Models, Molecular , Adaptor Proteins, Signal Transducing/physiology , Aminopeptidases/chemistry , Binding Sites , Foot-and-Mouth Disease Virus/genetics , Lysine/chemistry , Methionyl Aminopeptidases , Protein Biosynthesis , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/physiology , RNA, Small Interfering/genetics , RNA, Viral/chemistry , Transcriptional Activation
18.
Anal Biochem ; 368(2): 130-7, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17631855

ABSTRACT

Foot-and-mouth disease virus is a highly contagious pathogen that spreads rapidly among livestock and is capable of causing widespread agricultural and economic devastation. The virus genome is translated to produce a single polypeptide chain that subsequently is cleaved by viral proteases into mature protein products, with one protease, 3C(pro), carrying out the majority of the cleavages. The highly conserved nature of this protease across different viral strains and its crucial role in viral maturation and replication make it a very desirable target for inhibitor design. However, the lack of a convenient and high-throughput assay has been a hindrance in the characterization of potential inhibitors. In this article, we report the development of a continuous assay with potential for high throughput using fluorescence resonance energy transfer-based peptide substrates. Several peptide substrates containing the 3C-specific cleavage site were synthesized, varying both the positions and separation of the fluorescent donor and quencher groups. The best substrate, with a specificity constant k(cat)/K(M) of 57.6+/-2.0M(-1) s(-1), was used in inhibition assays to further characterize the protease's activity against a range of commercially available inhibitors. The inhibition profile of the enzyme showed characteristics of both cysteine and serine proteases, with the chymotrypsin inhibitor TPCK giving stoichiometric inhibition of the enzyme and allowing active site titration of the 3C(pro).


Subject(s)
Cysteine Endopeptidases/analysis , Fluorescence Resonance Energy Transfer/methods , Viral Proteins/analysis , 3C Viral Proteases , Cysteine Proteinase Inhibitors/pharmacology , Hydrolysis , Kinetics , Naphthalenesulfonates/chemistry , Naphthalenesulfonates/metabolism , Peptides/chemistry , Serine Proteinase Inhibitors/pharmacology , Substrate Specificity , Viral Proteins/antagonists & inhibitors , p-Dimethylaminoazobenzene/analogs & derivatives , p-Dimethylaminoazobenzene/chemistry , p-Dimethylaminoazobenzene/metabolism
19.
J Virol ; 81(1): 115-24, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17065215

ABSTRACT

The 3C protease (3C(pro)) from foot-and-mouth disease virus (FMDV), the causative agent of a widespread and economically devastating disease of domestic livestock, is a potential target for antiviral drug design. We have determined the structure of a new crystal form of FMDV 3C(pro), a chymotrypsin-like cysteine protease, which reveals features that are important for catalytic activity. In particular, we show that a surface loop which was disordered in previous structures adopts a beta-ribbon structure that is conformationally similar to equivalent regions on other picornaviral 3C proteases and some serine proteases. This beta-ribbon folds over the peptide binding cleft and clearly contributes to substrate recognition. Replacement of Cys142 at the tip of the beta-ribbon with different amino acids has a significant impact on enzyme activity and shows that higher activity is obtained with more hydrophobic side chains. Comparison of the structure of FMDV 3C(pro) with homologous enzyme-peptide complexes suggests that this correlation arises because the side chain of Cys142 contacts the hydrophobic portions of the P2 and P4 residues in the peptide substrate. Collectively, these findings provide compelling evidence for the role of the beta-ribbon in catalytic activity and provide valuable insights for the design of FMDV 3C(pro) inhibitors.


Subject(s)
Cysteine Endopeptidases/chemistry , Foot-and-Mouth Disease Virus/enzymology , Viral Proteins/chemistry , 3C Viral Proteases , Binding Sites , Crystallography, X-Ray , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Foot-and-Mouth Disease Virus/genetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary/physiology , Substrate Specificity , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 5): 646-50, 2005 May.
Article in English | MEDLINE | ID: mdl-15858279

ABSTRACT

Foot-and-mouth disease virus (FMDV) 3C protease (3C(pro)) plays a vital role in virus replication by performing most of the cleavages required to divide the viral polyprotein precursor into its functional component proteins. To date, no structural information has been available for FMDV 3C(pro), which is an attractive target for antiviral drugs. Targeted mutagenesis of surface amino acids identified two Cys residues that were detrimental to solubility and contributed to the time-dependent formation of a proteinaceous skin in samples of purified wild-type protein. Substitution of these amino acids, combined with trimming of the N- and C-termini, yielded a 3C(pro) construct that was amenable to crystallization. High-resolution diffraction (1.9 A) was only obtained following 'iterative screening' in which commercial crystal screening solutions were used as additives once initial crystallization conditions had been obtained.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , 3C Viral Proteases , Amino Acid Substitution , Crystallization , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Indicators and Reagents , Light , Models, Molecular , Mutagenesis , Plasmids , Scattering, Radiation , Solubility , Surface Properties , Thrombin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...