Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 15(6): 419, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879621

ABSTRACT

TRPV6 calcium channel is a prospective target in prostate cancer (PCa) since it is not expressed in healthy prostate while its expression increases during cancer progression. Despite the role of TRPV6 in PCa cell survival and apoptotic resistance has been already established, no reliable tool to target TRPV6 channel in vivo and thus to reduce tumor burden is known to date. Here we report the generation of mouse monoclonal antibody mAb82 raised against extracellular epitope of the pore region of the channel. mAb82 inhibited TRPV6 currents by 90% at 24 µg/ml in a dose-dependent manner while decreasing store-operated calcium entry to 56% at only 2.4 µg/ml. mAb82 decreased PCa survival rate in vitro by 71% at 12 µg/ml via inducing cell death through the apoptosis cascade via activation of the protease calpain, following bax activation, mitochondria enlargement, and loss of cristae, Cyt C release, pro-caspase 9 cleavage with the subsequent activation of caspases 3/7. In vivo, mice bearing either PC3Mtrpv6+/+ or PC3Mtrpv6-/-+pTRPV6 tumors were successfully treated with mAb82 at the dose as low as 100 µg/kg resulting in a significant reduction tumor growth by 31% and 90%, respectively. The survival rate was markedly improved by 3.5 times in mice treated with mAb82 in PC3Mtrpv6+/+ tumor group and completely restored in PC3Mtrpv6-/-+pTRPV6 tumor group. mAb82 showed a TRPV6-expression dependent organ distribution and virtually no toxicity in the same way as mAbAU1, a control antibody of the same Ig2a isotype. Overall, our data demonstrate for the first time the use of an anti-TRPV6 monoclonal antibody in vitro and in vivo in the treatment of the TRPV6-expressing PCa tumors.


Subject(s)
Antibodies, Monoclonal , Apoptosis , Calcium Channels , Prostatic Neoplasms , TRPV Cation Channels , Male , TRPV Cation Channels/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Apoptosis/drug effects , Humans , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Mice , Calcium Channels/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Calpain/metabolism , Calcium/metabolism
2.
Nat Commun ; 14(1): 5291, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37652913

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune, inflammatory and fibrotic disease with limited treatment options. Developing new therapies is therefore crucial to address patient needs. To this end, we focused on galectin-3 (Gal-3), a lectin known to be associated with several pathological processes seen in SSc. Using RNA sequencing of whole-blood samples in a cross-sectional cohort of 249 patients with SSc, Gal-3 and its interactants defined a strong transcriptomic fingerprint associated with disease severity, pulmonary and cardiac malfunctions, neutrophilia and lymphopenia. We developed new Gal-3 neutralizing monoclonal antibodies (mAb), which were then evaluated in a mouse model of hypochlorous acid (HOCl)-induced SSc. We show that two of these antibodies, D11 and E07, reduced pathological skin thickening, lung and skin collagen deposition, pulmonary macrophage content, and plasma interleukin-5 and -6 levels. Moreover, E07 changed the transcriptional profiles of HOCl-treated mice, resulting in a gene expression pattern that resembled that of control mice. Similarly, pathological pathways engaged in patients with SSc were counteracted by E07 in mice. Collectively, these findings demonstrate the translational potential of Gal-3 blockade as a therapeutic option for SSc.


Subject(s)
Galectin 3 , Scleroderma, Systemic , Animals , Mice , Galectin 3/genetics , Cross-Sectional Studies , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Antibodies, Monoclonal , Disease Models, Animal , Hypochlorous Acid
3.
J Immunother Cancer ; 7(1): 29, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30717773

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) treatment is currently restricted to chemotherapy. Hence, tumor-specific molecular targets and/or alternative therapeutic strategies for TNBC are urgently needed. Immunotherapy is emerging as an exciting treatment option for TNBC patients. The aspartic protease cathepsin D (cath-D), a marker of poor prognosis in breast cancer (BC), is overproduced and hypersecreted by human BC cells. This study explores whether cath-D is a tumor cell-associated extracellular biomarker and a potent target for antibody-based therapy in TNBC. METHODS: Cath-D prognostic value and localization was evaluated by transcriptomics, proteomics and immunohistochemistry in TNBC. First-in-class anti-cath-D human scFv fragments binding to both human and mouse cath-D were generated using phage display and cloned in the human IgG1 λ format (F1 and E2). Anti-cath-D antibody biodistribution, antitumor efficacy and in vivo underlying mechanisms were investigated in TNBC MDA-MB-231 tumor xenografts in nude mice. Antitumor effect was further assessed in TNBC patient-derived xenografts (PDXs). RESULTS: High CTSD mRNA levels correlated with shorter recurrence-free survival in TNBC, and extracellular cath-D was detected in the tumor microenvironment, but not in matched normal breast stroma. Anti-cath-D F1 and E2 antibodies accumulated in TNBC MDA-MB-231 tumor xenografts, inhibited tumor growth and improved mice survival without apparent toxicity. The Fc function of F1, the best antibody candidate, was essential for maximal tumor inhibition in the MDA-MB-231 model. Mechanistically, F1 antitumor response was triggered through natural killer cell activation via IL-15 upregulation, associated with granzyme B and perforin production, and the release of antitumor IFNγ cytokine. The F1 antibody also prevented the tumor recruitment of immunosuppressive tumor-associated macrophages M2 and myeloid-derived suppressor cells, a specific effect associated with a less immunosuppressive tumor microenvironment highlighted by TGFß decrease. Finally, the antibody F1 inhibited tumor growth of two TNBC PDXs, isolated from patients resistant or not to neo-adjuvant chemotherapy. CONCLUSION: Cath-D is a tumor-specific extracellular target in TNBC suitable for antibody-based therapy. Immunomodulatory antibody-based strategy against cath-D is a promising immunotherapy to treat patients with TNBC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cathepsin D/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Cathepsin D/genetics , Cathepsin D/immunology , Cell Line, Tumor , Female , Humans , Immunotherapy , Mice, Nude , RNA, Messenger/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Methods Mol Biol ; 1827: 93-108, 2018.
Article in English | MEDLINE | ID: mdl-30196493

ABSTRACT

Libraries of antibody fragments displayed on filamentous phages are now a widely used approach to isolate antibodies against virtually any target. We describe a simple protocol to make large and diverse libraries based on a single or a limited number of frameworks. The approach is flexible enough to be used with any antibody format, either single-chain (scFv, VHH) or multi-chain (Fv, Fab, (Fab')2), and to target in a single step the six complementarity-determining regions-or any other part-of the antibody molecule. Using this protocol, libraries larger than 1010 can be constructed in a single week.


Subject(s)
Peptide Library , Protein Engineering/methods , DNA/metabolism , Electroporation , Escherichia coli/metabolism , Mutagenesis/genetics , Quality Control
5.
Methods Mol Biol ; 1701: 239-253, 2018.
Article in English | MEDLINE | ID: mdl-29116508

ABSTRACT

Libraries of antibody fragments displayed on filamentous phages have proved their value to generate human antibodies against virtually any target. We describe here a simple protocol to make large and diverse libraries based on a single or a limited number of frameworks. The approach is flexible enough to be used with any antibody format, either single-chain (scFv, VHH) or multi-chain (Fv, Fab, (Fab')2), and to target in a single step the six complementarity-determining regions-or any other part-of the antibody molecule. Using this protocol, libraries larger than 1010 can be easily constructed in a single week.


Subject(s)
Cloning, Molecular/methods , Gene Library , Peptide Library , Single-Chain Antibodies/genetics , Animals , Humans , Single-Chain Antibodies/immunology
7.
J Mol Biol ; 426(22): 3729-3743, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25174334

ABSTRACT

Antibody molecules are able to recognize any antigen with high affinity and specificity. To get insight into the molecular diversity at the source of this functional diversity, we compiled and analyzed a non-redundant aligned collection of 227 structures of antibody-antigen complexes. Free energy of binding of all the residue side chains was quantified by computational alanine scanning, allowing the first large-scale quantitative description of antibody paratopes. This demonstrated that as few as 8 residues among 30 key positions are sufficient to explain 80% of the binding free energy in most complexes. At these positions, the residue distribution is not only different from that of other surface residues but also dependent on the role played by the side chain in the interaction, residues participating in the binding energy being mainly aromatic residues, and Gly or Ser otherwise. To question the generality of these binding characteristics, we isolated an antibody fragment by phage display using a biased synthetic repertoire with only two diversified complementarity-determining regions and solved its structure in complex with its antigen. Despite this restricted diversity, the structure demonstrated that all complementarity-determining regions were involved in the interaction with the antigen and that the rules derived from the natural antibody repertoire apply to this synthetic binder, thus demonstrating the robustness and universality of our results.


Subject(s)
Alanine/chemistry , Antibodies/chemistry , Antibodies/metabolism , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Complementarity Determining Regions/chemistry , Alanine/genetics , Alanine/metabolism , Antibodies/genetics , Antigen-Antibody Complex/genetics , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Humans , Hydrogen Bonding , Models, Molecular , Mutagenesis , Mutation/genetics , Peptide Library , Protein Binding , Protein Conformation
8.
J Mol Recognit ; 27(9): 549-58, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25042709

ABSTRACT

Intrabodies, when expressed in cells after genetic fusion to fluorescent proteins, are powerful tools to study endogenous protein dynamics inside cells. However, it remains challenging to determine the conditions for specific imaging and precise labelling of the target antigen with such intracellularly expressed antibody fragments. Here, we show that single-chain Fv (scFv) antibody fragments can be generated that specifically recognize proliferating cell nuclear antigen (PCNA) when expressed in living cancer cells. After selection by phage display, the anti-PCNA scFvs were screened in vitro after being tagged with dimeric glutathione-S-transferase. Anti-PCNA scFvs of increased avidity were further engineered by mutagenesis with sodium bisulfite and error-prone PCR, such that they were almost equivalent to conventional antibodies in in vitro assays. These intrabodies were then rendered bifunctional by fusion to a C-terminal fragment of p21 protein and could thereby readily detect PCNA bound to chromatin in cells. Finally, by linking these optimized peptide-conjugated scFvs to an enhanced green fluorescent protein, fluorescent intrabody-based reagents were obtained that allowed the fate of PCNA in living cells to be examined. The approach described may be applicable to other scFvs that can be solubly expressed in cells, and it provides a unique means to recognize endogenous proteins in living cells with high accuracy.


Subject(s)
Diagnostic Imaging , Neoplasms/diagnosis , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Sequence , Antibody Affinity , Cell Line, Tumor , Cell Survival , Fluorescence , Humans , Indicators and Reagents , Molecular Sequence Data , Peptide Library , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Subcellular Fractions/metabolism
9.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 774-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23633586

ABSTRACT

The caspase recruitment domain (CARD) is present in death-domain superfamily proteins involved in inflammation and apoptosis. BinCARD is named for its ability to interact with Bcl10 and inhibit downstream signalling. Human BinCARD is expressed as two isoforms that encode the same N-terminal CARD region but which differ considerably in their C-termini. Both isoforms are expressed in immune cells, although BinCARD-2 is much more highly expressed. Crystals of the CARD fold common to both had low symmetry (space group P1). Molecular replacement was unsuccessful in this low-symmetry space group and, as the construct contains no methionines, first one and then two residues were engineered to methionine for MAD phasing. The double-methionine variant was produced as a selenomethionine derivative, which was crystallized and the structure was solved using data measured at two wavelengths. The crystal structures of the native and selenomethionine double mutant were refined to high resolution (1.58 and 1.40 Šresolution, respectively), revealing the presence of a cis-peptide bond between Tyr39 and Pro40. Unexpectedly, the native crystal structure revealed that all three cysteines were oxidized. The mitochondrial localization of BinCARD-2 and the susceptibility of its CARD region to redox modification points to the intriguing possibility of a redox-regulatory role.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , Proteins/chemistry , Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Crystallography, X-Ray , Cysteine/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Models, Molecular , Mutation , Oxidation-Reduction , Proline/chemistry , Protein Conformation , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/genetics , Selenomethionine
10.
Methods Mol Biol ; 907: 109-22, 2012.
Article in English | MEDLINE | ID: mdl-22907348

ABSTRACT

Libraries of antibody fragments displayed on filamentous phages have proved their value to generate human antibodies against virtually any target. We describe here a simple protocol to make large and diverse libraries based on a single or few frameworks. Diversity is introduced in the third hypervariable loops using degenerate synthetic oligonucleotides and PCR assembly. Because all the antibody fragments isolated from the library will share their framework sequence, their stability and physical properties will be more consistent and customizable than when antibody fragments are isolated from a library prepared from human donors.


Subject(s)
Molecular Biology/methods , Peptide Library , Single-Chain Antibodies/genetics , Bacteriophages/metabolism , Cloning, Molecular , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Electroporation , Genetic Vectors/genetics , Humans , Oligonucleotides , Protein Structure, Secondary , Single-Chain Antibodies/chemistry , Transformation, Genetic
11.
J Biol Chem ; 286(28): 25397-405, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21622577

ABSTRACT

The SPP1 siphophage uses its long non-contractile tail and tail tip to recognize and infect the Gram-positive bacterium Bacillus subtilis. The tail-end cap and its attached tip are the critical components for host recognition and opening of the tail tube for genome exit. In the present work, we determined the cryo-electron microscopic (cryo-EM) structure of a complex formed by the cap protein gp19.1 (Dit) and the N terminus of the downstream protein of gp19.1 in the SPP1 genome, gp21(1-552) (Tal). This complex assembles two back-to-back stacked gp19.1 ring hexamers, interacting loosely, and two gp21(1-552) trimers interacting with gp19.1 at both ends of the stack. Remarkably, one gp21(1-552) trimer displays a "closed" conformation, whereas the second is "open" delineating a central channel. The two conformational states dock nicely into the EM map of the SPP1 cap domain, respectively, before and after DNA release. Moreover, the open/closed conformations of gp19.1-gp21(1-552) are consistent with the structures of the corresponding proteins in the siphophage p2 baseplate, where the Tal protein (ORF16) attached to the ring of Dit (ORF15) was also found to adopt these two conformations. Therefore, the present contribution allowed us to revisit the SPP1 tail distal-end architectural organization. Considering the sequence conservation among Dit and the N-terminal region of Tal-like proteins in Gram-positive-infecting Siphoviridae, it also reveals the Tal opening mechanism as a hallmark of siphophages probably involved in the generation of the firing signal initiating the cascade of events that lead to phage DNA release in vivo.


Subject(s)
Bacillus subtilis/virology , Genome, Viral/physiology , Siphoviridae/physiology , Viral Structural Proteins/metabolism , Virus Attachment , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacillus subtilis/ultrastructure , Protein Structure, Tertiary , Siphoviridae/ultrastructure , Viral Structural Proteins/genetics
12.
J Biol Chem ; 285(47): 36666-73, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20843802

ABSTRACT

Siphophage SPP1 infects the gram-positive bacterium Bacillus subtilis using its long non-contractile tail and tail-tip. Electron microscopy (EM) previously allowed a low resolution assignment of most orf products belonging to these regions. We report here the structure of the SPP1 distal tail protein (Dit, gp19.1). The combination of x-ray crystallography, EM, and light scattering established that Dit is a back-to-back dimer of hexamers. However, Dit fitting in the virion EM maps was only possible with a hexamer located between the tail-tube and the tail-tip. Structure comparison revealed high similarity between Dit and a central component of lactophage baseplates. Sequence similarity search expanded its relatedness to several phage proteins, suggesting that Dit is a docking platform for the tail adsorption apparatus in Siphoviridae infecting gram-positive bacteria and that its architecture is a paradigm for these hub proteins. Dit structural similarity extends also to non-contractile and contractile phage tail proteins (gpV(N) and XkdM) as well as to components of the bacterial type 6 secretion system, supporting an evolutionary connection between all these devices.


Subject(s)
Bacillus subtilis/virology , Bacteriophages/metabolism , Siphoviridae/genetics , Viral Tail Proteins/chemistry , Bacteriophages/genetics , Crystallography, X-Ray , Protein Conformation , Viral Regulatory and Accessory Proteins , Viral Tail Proteins/genetics , Viral Tail Proteins/metabolism , Virion/metabolism
13.
PLoS One ; 4(11): e7851, 2009 Nov 16.
Article in English | MEDLINE | ID: mdl-19924245

ABSTRACT

BACKGROUND: Protein crystallisation screening involves the parallel testing of large numbers of candidate conditions with the aim of identifying conditions suitable as a starting point for the production of diffraction quality crystals. Generally, condition screening is performed in 96-well plates. While previous studies have examined the effects of protein construct, protein purity, or crystallisation condition ingredients on protein crystallisation, few have examined the effect of the crystallisation plate. METHODOLOGY/PRINCIPAL FINDINGS: We performed a statistically rigorous examination of protein crystallisation, and evaluated interactions between crystallisation success and plate row/column, different plates of same make, different plate makes and different proteins. From our analysis of protein crystallisation, we found a significant interaction between plate make and the specific protein being crystallised. CONCLUSIONS/SIGNIFICANCE: Protein crystal structure determination is the principal method for determining protein structure but is limited by the need to produce crystals of the protein under study. Many important proteins are difficult to crystallize, so that identification of factors that assist crystallisation could open up the structure determination of these more challenging targets. Our findings suggest that protein crystallisation success may be improved by matching a protein with its optimal plate make.


Subject(s)
Crystallization/methods , Proteins/chemistry , Animals , Antigens/chemistry , Catalase/chemistry , Cattle , Chickens , Crystallography, X-Ray/instrumentation , Crystallography, X-Ray/methods , Egg White/chemistry , Escherichia coli/metabolism , Humans , Hydrogen-Ion Concentration , Liver/enzymology , Mice , Muramidase/chemistry , Pilot Projects , Streptomyces/metabolism
14.
J Biol Chem ; 284(34): 22549-58, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19549784

ABSTRACT

Ran-GTP interacts strongly with importin-beta, and this interaction promotes the release of the importin-alpha-nuclear localization signal cargo from importin-beta. Ran-GDP also interacts with importin-beta, but this interaction is 4 orders of magnitude weaker than the Ran-GTP.importin-beta interaction. Here we use the yeast complement of nuclear import proteins to show that the interaction between Ran-GDP and importin-beta promotes the dissociation of GDP from Ran. The release of GDP from the Ran-GDP-importin-beta complex stabilizes the complex, which cannot be dissociated by importin-alpha. Although Ran has a higher affinity for GDP compared with GTP, Ran in complex with importin-beta has a higher affinity for GTP. This feature is responsible for the generation of Ran-GTP from Ran-GDP by importin-beta. Ran-binding protein-1 (RanBP1) activates this reaction by forming a trimeric complex with Ran-GDP and importin-beta. Importin-alpha inhibits the GDP exchange reaction by sequestering importin-beta, whereas RanBP1 restores the GDP nucleotide exchange by importin-beta by forming a tetrameric complex with importin-beta, Ran, and importin-alpha. The exchange is also inhibited by nuclear-transport factor-2 (NTF2). We suggest a mechanism for nuclear import, additional to the established RCC1 (Ran-guanine exchange factor)-dependent pathway that incorporates these results.


Subject(s)
Active Transport, Cell Nucleus/physiology , Guanine Nucleotide Exchange Factors/metabolism , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/genetics , Chromatography, Gel , Chromatography, High Pressure Liquid , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding , alpha Karyopherins/metabolism
15.
Protein Expr Purif ; 67(2): 88-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19460440

ABSTRACT

Developmentally regulated G-proteins (DRGs) are a highly conserved family of GTP-binding proteins found in archaea, plants, fungi and animals, indicating important roles in fundamental pathways. Their function is poorly understood, but they have been implicated in cell division, proliferation, and growth, as well as several medical conditions. Individual subfamilies within the G-protein superfamily possess unique nucleotide binding and hydrolysis rates that are intrinsic to their cellular function, and so characterization of these rates for a particular G-protein may provide insight into its cellular activity. We have produced recombinant active DRG protein using a bacterial expression system and refolding, and performed biochemical characterization of their GTP binding and hydrolysis. We show that recombinant Arabidopsis thaliana atDRG1 and atDRG2a are able to bind GDP and GTP. We also show that DRGs can hydrolyze GTP in vitro without the assistance of GTPase-activating proteins and guanine exchange factors. The atDRG proteins hydrolyze GTP at a relatively slow rate (0.94x10(-3)min(-1) for DRG1 and 1.36x10(-3)min(-1) for DRG2) that is consistent with their nearest characterized relatives, the Obg subfamily. The ability of DRGs to bind nucleotide substrates without assistance, their slow rate of GTP hydrolysis, heat stress activation and domain conservation suggest a possible role as a chaperone in ribosome assembly in response to stress as it has been suggested for the Obg proteins, a different but related G-protein subfamily.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , GTP-Binding Proteins/chemistry , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Escherichia coli/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Inclusion Bodies/chemistry , Mass Spectrometry , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , ortho-Aminobenzoates/metabolism
16.
J Mol Biol ; 385(5): 1568-77, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19059417

ABSTRACT

Over the last decade, West Nile virus has spread rapidly via mosquito transmission from infected migratory birds to humans. One potential therapeutic approach to treating infection is to inhibit the virally encoded serine protease that is essential for viral replication. Here we report the crystal structure of the viral NS3 protease tethered to its essential NS2B cofactor and bound to a potent substrate-based tripeptide inhibitor, 2-naphthoyl-Lys-Lys-Arg-H (K(i)=41 nM), capped at the N-terminus by 2-naphthoyl and capped at the C-terminus by aldehyde. An important and unexpected feature of this structure is the presence of two conformations of the catalytic histidine suggesting a role for ligand stabilization of the catalytically competent His conformation. Analysis of other West Nile virus NS3 protease structures and related serine proteases supports this hypothesis, suggesting that the common catalytic mechanism involves an induced-fit mechanism.


Subject(s)
Models, Molecular , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry , West Nile virus/enzymology , Amino Acid Sequence , Catalysis , Catalytic Domain , Ligands , Molecular Sequence Data , Protein Conformation , RNA Helicases/antagonists & inhibitors , RNA Helicases/chemistry , Serine Proteinase Inhibitors/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , X-Ray Diffraction
17.
J Mol Biol ; 383(4): 772-82, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18708071

ABSTRACT

The asymmetric distribution of the nucleotide-bound state of Ran across the nuclear envelope is crucial for determining the directionality of nuclear transport. In the nucleus, Ran is primarily in the guanosine 5'-triphosphate (GTP)-bound state, whereas in the cytoplasm, Ran is primarily guanosine 5'-diphosphate (GDP)-bound. Conformational changes within the Ran switch I and switch II loops are thought to modulate its affinity for importin-beta. Here, we show that RanGDP and importin-beta form a stable complex with a micromolar dissociation constant. This complex can be dissociated by importin-beta binding partners such as importin-alpha. Surprisingly, the crystal structure of the Kap95p-RanGDP complex shows that Kap95p induces the switch I and II regions of RanGDP to adopt a conformation that resembles that of the GTP-bound form. The structure of the complex provides insights into the structural basis for the gradation of affinities regulating nuclear protein transport.


Subject(s)
Active Transport, Cell Nucleus/physiology , Guanosine Diphosphate/metabolism , Protein Conformation , beta Karyopherins/chemistry , beta Karyopherins/metabolism , ran GTP-Binding Protein/chemistry , ran GTP-Binding Protein/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics
18.
Methods Mol Biol ; 426: 27-35, 2008.
Article in English | MEDLINE | ID: mdl-18542855

ABSTRACT

Increasing the success in obtaining structures and maximizing the value of the structures determined are the two major goals of target selection in structural proteomics. This chapter presents an efficient and flexible target selection procedure supplemented with a Web-based resource that is suitable for small- to large-scale structural genomics projects that use crystallography as the major means of structure determination. Based on three criteria, biological significance, structural novelty, and "crystallizability," the approach first removes (filters) targets that do not meet minimal criteria and then ranks the remaining targets based on their "crystallizability" estimates. This novel procedure was designed to maximize selection efficiency, and its prevailing criteria categories make it suitable for a broad range of structural proteomics projects.


Subject(s)
Proteins/chemistry , Structural Homology, Protein , Animals , Crystallization , Crystallography, X-Ray/methods , Databases, Protein , Humans
19.
Methods Mol Biol ; 426: 269-75, 2008.
Article in English | MEDLINE | ID: mdl-18542870

ABSTRACT

Expression of insoluble protein in E. coli is a major bottleneck of high throughput structural biology projects. Refolding proteins into native conformations from inclusion bodies could significantly increase the number of protein targets that can be taken on to structural studies. This chapter presents a simple assay for screening insoluble protein targets and identifying those that are most amenable to refolding. The assay is based on the observation that when proteins are refolded while bound to metal affinity resin, misfolded proteins are generally not eluted by imidazole. This difference is exploited here to distinguish between folded and misfolded proteins. Two implementations of the assay are described. The assay fits well into a standard high throughput structural biology pipeline, because it begins with the inclusion body preparations that are a byproduct of small-scale, automated expression and purification trials and does not require additional facilities. Two formats of the assay are described, a manual assay that is useful for screening small numbers of targets, and an automated implementation that is useful for large numbers of targets.


Subject(s)
Protein Folding , Protein Renaturation , Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Histidine/chemistry , Inclusion Bodies/chemistry , Proteins/genetics , Proteins/isolation & purification , Solubility
20.
Methods Mol Biol ; 426: 577-87, 2008.
Article in English | MEDLINE | ID: mdl-18542891

ABSTRACT

This chapter describes the methodology adopted in a project aimed at structural and functional characterization of proteins that potentially play an important role in mammalian macrophages. The methodology that underpins this project is applicable to both small research groups and larger structural genomics consortia. Gene products with putative roles in macrophage function are identified using gene expression information obtained via DNA microarray technology. Specific targets for structural and functional characterization are then selected based on a set of criteria aimed at maximizing insight into function. The target proteins are cloned using a modification of Gateway cloning technology, expressed with hexa-histidine tags in E. coli, and purified to homogeneity using a combination of affinity and size exclusion chromatography. Purified proteins are finally subjected to crystallization trials and/or NMR-based screening to identify candidates for structure determination. Where crystallography and NMR approaches are unsuccessful, chemical cross-linking is employed to obtain structural information. This resulting structural information is used to guide cell biology experiments to further investigate the cellular and molecular function of the targets in macrophage biology. Jointly, the data sheds light on the molecular and cellular functions of macrophage proteins.


Subject(s)
Macrophages/metabolism , Proteins/chemistry , Proteomics/methods , Proteomics/organization & administration , Animals , Arthritis/genetics , Arthritis/metabolism , Computational Biology , Crystallography, X-Ray , Humans , Mice , Protein Conformation , Protein Folding , Proteins/genetics , Proteins/isolation & purification , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Queensland , Universities
SELECTION OF CITATIONS
SEARCH DETAIL