Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
STAR Protoc ; 4(4): 102688, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37943662

ABSTRACT

Differential scanning fluorimetry (DSF) is a widely used technique for determining the apparent melting temperature (Tma) of a purified protein. Here, we present a protocol for performing and optimizing DSF experiments. We describe steps for designing and performing the experiment, analyzing data, and optimization. We provide benchmarks for typical Tmas and ΔTmas, standard assay conditions, and upper and lower limits of commonly altered experimental variables. We also detail common pitfalls of DSF and ways to avoid, identify, and overcome them.


Subject(s)
Amines , Proteins , Calorimetry, Differential Scanning , Temperature , Fluorometry/methods
2.
Anal Chem ; 93(49): 16474-16480, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34854675

ABSTRACT

Identification of antibodies targeting diverse functional epitopes on an antigen is highly crucial for discovering effective therapeutic candidates. Employing a traditional stepwise antibody "screening funnel" as well as prioritizing affinity-based selections over epitope-based selections, result in lead antibody panels lacking epitope diversity. In the present study, we employed an array-based surface plasmon resonance (SPR) platform to perform high-throughput epitope binning analysis on a large number of monoclonal antibodies (mAbs) generated in the early drug discovery process. The mAb panel contained clones from different antibody generation techniques and diverse transgenic mouse strains. The epitope binning results were analyzed in unique ways using various visualizations in the form of dendrograms and network plots, which assisted in determining diversity and redundancy in the mAb sample set. The binning data were further integrated with affinity information to evaluate the performance of seven different transgenic mouse strains. The combination of epitope binning results with binding kinetics and sequence analysis provided an effective and efficient way of selecting high affinity antibodies representing a diverse set of sequence families and epitopes.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents, Immunological , Animals , Epitopes , Mice , Surface Plasmon Resonance
3.
Proc Natl Acad Sci U S A ; 115(11): 2836-2841, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29476010

ABSTRACT

Human cells express thousands of different surface proteins that can be used for cell classification, or to distinguish healthy and disease conditions. A method capable of profiling a substantial fraction of the surface proteome simultaneously and inexpensively would enable more accurate and complete classification of cell states. We present a highly multiplexed and quantitative surface proteomic method using genetically barcoded antibodies called phage-antibody next-generation sequencing (PhaNGS). Using 144 preselected antibodies displayed on filamentous phage (Fab-phage) against 44 receptor targets, we assess changes in B cell surface proteins after the development of drug resistance in a patient with acute lymphoblastic leukemia (ALL) and in adaptation to oncogene expression in a Myc-inducible Burkitt lymphoma model. We further show PhaNGS can be applied at the single-cell level. Our results reveal that a common set of proteins including FLT3, NCR3LG1, and ROR1 dominate the response to similar oncogenic perturbations in B cells. Linking high-affinity, selective, genetically encoded binders to NGS enables direct and highly multiplexed protein detection, comparable to RNA-sequencing for mRNA. PhaNGS has the potential to profile a substantial fraction of the surface proteome simultaneously and inexpensively to enable more accurate and complete classification of cell states.


Subject(s)
Antibodies/analysis , Burkitt Lymphoma/genetics , High-Throughput Nucleotide Sequencing/methods , Leukemia/genetics , Membrane Proteins/genetics , Proteomics/methods , Antibodies/genetics , Bacteriophages/genetics , Bacteriophages/metabolism , Burkitt Lymphoma/metabolism , Cell Line, Tumor , Humans , Leukemia/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism
4.
J Mol Biol ; 429(23): 3666-3677, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28942089

ABSTRACT

Heterochromatin protein 1 (HP1) family proteins are conserved chromatin binding proteins involved in gene silencing, chromosome packaging, and chromosome segregation. These proteins recognize histone H3 lysine 9 methylated tails via their chromodomain and recruit additional ligand proteins with diverse activities through their dimerization domain, the chromoshadow domain. Species that have HP1 proteins possess multiple paralogs that perform non-overlapping roles in vivo. How different HP1 proteins, which are highly conserved, perform different functions is not well understood. Here, we use the two Schizosaccharomyces pombe HP1 paralogs, Swi6 and Chp2, as model systems to compare and contrast their biophysical properties. We find that Swi6 and Chp2 have similar dimerization and oligomerization equilibria, and that Swi6 binds slightly (~3-fold) more strongly to nucleosomes than Chp2. Furthermore, while Swi6 binding to the H3K9me3 mark is regulated by a previously described auto-inhibition mechanism, the binding of Chp2 to the H3K9me3 mark is not analogously regulated. In the context of chromoshadow domain interactions, we show using a newly identified peptide sequence from the Clr3 histone deacetylase and a previously identified sequence from the protein Shugoshin that the Swi6 chromoshadow domain binds both ligands more strongly than the Chp2. Overall, our findings uncover quantitative differences in how Swi6 and Chp2 interact with nucleosomal and non-nucleosomal ligands and qualitative differences in how their assembly on nucleosomes is regulated. These findings provide a biochemical framework to explain the varied functions of Chp2 and Swi6 in vivo.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Nucleosomes/metabolism , Repressor Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Heterochromatin/genetics , Protein Conformation , Repressor Proteins/chemistry , Repressor Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics
5.
Science ; 355(6325): 597-602, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183972

ABSTRACT

Cysteine can be specifically functionalized by a myriad of acid-base conjugation strategies for applications ranging from probing protein function to antibody-drug conjugates and proteomics. In contrast, selective ligation to the other sulfur-containing amino acid, methionine, has been precluded by its intrinsically weaker nucleophilicity. Here, we report a strategy for chemoselective methionine bioconjugation through redox reactivity, using oxaziridine-based reagents to achieve highly selective, rapid, and robust methionine labeling under a range of biocompatible reaction conditions. We highlight the broad utility of this conjugation method to enable precise addition of payloads to proteins, synthesis of antibody-drug conjugates, and identification of hyperreactive methionine residues in whole proteomes.


Subject(s)
Aziridines/chemistry , Cysteine/chemistry , Immunoconjugates/chemistry , Methionine/chemistry , Actins/chemistry , Gene Editing , Gene Knockout Techniques , Methionine/analysis , Mutation , Oxidation-Reduction , Phosphopyruvate Hydratase/genetics , Protein Domains , Proteins/chemistry , Proteomics/methods , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Sodium Hypochlorite/pharmacology
6.
Sci Rep ; 6: 39774, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008969

ABSTRACT

High-quality immunoreagents enhance the performance and reproducibility of immunoassays and, in turn, the quality of both biological and clinical measurements. High quality recombinant immunoreagents are generated using antibody-phage display. One metric of antibody quality - the binding affinity - is quantified through the dissociation constant (KD) of each recombinant antibody and the target antigen. To characterize the KD of recombinant antibodies and target antigen, we introduce affinity electrophoretic mobility shift assays (EMSAs) in a high-throughput format suitable for small volume samples. A microfluidic card comprised of free-standing polyacrylamide gel (fsPAG) separation lanes supports 384 concurrent EMSAs in 30 s using a single power source. Sample is dispensed onto the microfluidic EMSA card by acoustic droplet ejection (ADE), which reduces EMSA variability compared to sample dispensing using manual or pin tools. The KD for each of a six-member fragment antigen-binding fragment library is reported using ~25-fold less sample mass and ~5-fold less time than conventional heterogeneous assays. Given the form factor and performance of this micro- and mesofluidic workflow, we have developed a sample-sparing, high-throughput, solution-phase alternative for biomolecular affinity characterization.


Subject(s)
Antibody Affinity , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Immunoglobulin Fab Fragments/chemistry , Animals , Humans , Recombinant Proteins/chemistry
7.
Mol Cell Proteomics ; 14(10): 2833-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290498

ABSTRACT

Antibodies are key reagents in biology and medicine, but commercial sources are rarely recombinant and thus do not provide a permanent and renewable resource. Here, we describe an industrialized platform to generate antigens and validated recombinant antibodies for 346 transcription factors (TFs) and 211 epigenetic antigens. We describe an optimized automated phage display and antigen expression pipeline that in aggregate produced about 3000 sequenced Fragment antigen-binding domain that had high affinity (typically EC50<20 nm), high stability (Tm∼80 °C), good expression in E. coli (∼5 mg/L), and ability to bind antigen in complex cell lysates. We evaluated a subset of Fabs generated to homologous SCAN domains for binding specificities. These Fragment antigen-binding domains were monospecific to their target SCAN antigen except in rare cases where they cross-reacted with a few highly related antigens. Remarkably, immunofluorescence experiments in six cell lines for 270 of the TF antigens, each having multiple antibodies, show that ∼70% stain predominantly in the cytosol and ∼20% stain in the nucleus which reinforces the dominant role that translocation plays in TF biology. These cloned antibody reagents are being made available to the academic community through our web site recombinant-antibodies.org to allow a more system-wide analysis of TF and chromatin biology. We believe these platforms, infrastructure, and automated approaches will facilitate the next generation of renewable antibody reagents to the human proteome in the coming decade.


Subject(s)
Antibodies , Immunoglobulin Fab Fragments , Transcription Factors , Antibodies/genetics , Antibodies/immunology , Antigens/genetics , Antigens/immunology , Escherichia coli/genetics , High-Throughput Screening Assays , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Protein Folding , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transcription Factors/genetics , Transcription Factors/immunology
8.
J Vis Exp ; (95): 51492, 2015 Jan 17.
Article in English | MEDLINE | ID: mdl-25651360

ABSTRACT

The demand for antibodies that fulfill the needs of both basic and clinical research applications is high and will dramatically increase in the future. However, it is apparent that traditional monoclonal technologies are not alone up to this task. This has led to the development of alternate methods to satisfy the demand for high quality and renewable affinity reagents to all accessible elements of the proteome. Toward this end, high throughput methods for conducting selections from phage-displayed synthetic antibody libraries have been devised for applications involving diverse antigens and optimized for rapid throughput and success. Herein, a protocol is described in detail that illustrates with video demonstration the parallel selection of Fab-phage clones from high diversity libraries against hundreds of targets using either a manual 96 channel liquid handler or automated robotics system. Using this protocol, a single user can generate hundreds of antigens, select antibodies to them in parallel and validate antibody binding within 6-8 weeks. Highlighted are: i) a viable antigen format, ii) pre-selection antigen characterization, iii) critical steps that influence the selection of specific and high affinity clones, and iv) ways of monitoring selection effectiveness and early stage antibody clone characterization. With this approach, we have obtained synthetic antibody fragments (Fabs) to many target classes including single-pass membrane receptors, secreted protein hormones, and multi-domain intracellular proteins. These fragments are readily converted to full-length antibodies and have been validated to exhibit high affinity and specificity. Further, they have been demonstrated to be functional in a variety of standard immunoassays including Western blotting, ELISA, cellular immunofluorescence, immunoprecipitation and related assays. This methodology will accelerate antibody discovery and ultimately bring us closer to realizing the goal of generating renewable, high quality antibodies to the proteome.


Subject(s)
Antibodies/immunology , Antigens/immunology , High-Throughput Screening Assays/methods , Peptide Library , Antigen-Antibody Reactions , Enzyme-Linked Immunosorbent Assay/methods , Epitopes , Humans , Immunoassay/methods
9.
J Mol Biol ; 427(2): 576-86, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25481745

ABSTRACT

Antibody phage display libraries combined with high-throughput selections have recently demonstrated tremendous promise to create the next generation of renewable, recombinant antibodies to study proteins and their many post-translational modification states; however, many challenges still remain, such as optimized antibody scaffolds. Recently, a single-chain fragment antigen binding (Fab) (scFab) format, in which the carboxy-terminus of the light chain is linked to the amino-terminus of the heavy chain, was described to potentially combine the high display levels of a single-chain fragment variable with the high stability of purified Fabs. However, this format required removal of the interchain disulfide bond to achieve modest display levels and subsequent bacterial expression resulted in high levels of aggregated scFab, hindering further use of scFabs. Here, we developed an improved scFab format that retains the interchain disulfide bond by increasing the linker length between the light and heavy chains to improve display and bacterial expression levels to 1-3 mg/L. Furthermore, rerouting of the scFab to the co-translational signal recognition particle pathway combined with reengineering of the signal peptide sequence results in display levels 24-fold above the original scFab format and 3-fold above parent Fab levels. This optimized scFab scaffold can be easily reformatted in a single step for expression in a bacterial or mammalian host to produce stable (Tm of 81 °C), predominantly monomeric (>90%) antibodies at a high yield. Ultimately, this new scFab format will advance high-throughput antibody generation platforms to discover the next generation of research and therapeutic antibodies.


Subject(s)
Computer Simulation , Gene Expression , Immunoglobulin Fab Fragments/chemistry , Recombinant Proteins/chemistry , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , Bacteria/genetics , Bacteriophages/genetics , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Mammals/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Protein Conformation , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Sequence Analysis, DNA , Single-Chain Antibodies/genetics
10.
Genome Biol Evol ; 6(11): 3025-38, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25349268

ABSTRACT

ß-defensins are a family of important peptides of innate immunity, involved in host defense, immunomodulation, reproduction, and pigmentation. Genes encoding ß-defensins show evidence of birth-and-death evolution, adaptation by amino acid sequence changes, and extensive copy number variation (CNV) within humans and other species. The role of CNV in the adaptation of ß-defensins to new functions remains unclear, as does the adaptive role of CNV in general. Here, we fine-map CNV of a cluster of ß-defensins in humans and rhesus macaques. Remarkably, we found that the structure of the CNV is different between primates, with distinct mutational origins and CNV boundaries defined by retroviral long terminal repeat elements. Although the human ß-defensin CNV region is 322 kb and encompasses several genes, including ß-defensins, a long noncoding RNA gene, and testes-specific zinc-finger transcription factors, the orthologous region in the rhesus macaque shows CNV of a 20-kb region, containing only a single gene, the ortholog of the human ß-defensin-2 gene. Despite its independent origins, the range of gene copy numbers in the rhesus macaque is similar to humans. In addition, the rhesus macaque gene has been subject to divergent positive selection at the amino acid level following its initial duplication event between 3 and 9.5 Ma, suggesting adaptation of this gene as the macaque successfully colonized novel environments outside Africa. Therefore, the molecular phenotype of ß-defensin-2 CNV has undergone convergent evolution, and this gene shows evidence of adaptation at the amino acid level in rhesus macaques.


Subject(s)
Adaptation, Physiological/genetics , DNA Copy Number Variations , Evolution, Molecular , beta-Defensins/genetics , Animals , Humans , Macaca mulatta , Mutation , Selection, Genetic
11.
Cell Host Microbe ; 5(5): 476-86, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19454351

ABSTRACT

In response to enteric pathogens, the inflamed intestine produces antimicrobial proteins, a process mediated by the cytokines IL-17 and IL-22. Salmonella enterica serotype Typhimurium thrives in the inflamed intestinal environment, suggesting that the pathogen is resistant to antimicrobials it encounters in the intestinal lumen. However, the identity of these antimicrobials and corresponding bacterial resistance mechanisms remain unknown. Here, we report that enteric infection of rhesus macaques and mice with S. Typhimurium resulted in marked Il-17- and IL-22-dependent intestinal epithelial induction and luminal accumulation of lipocalin-2, an antimicrobial protein that prevents bacterial iron acquisition. Resistance to lipocalin-2, mediated by the iroBCDE iroN locus, conferred a competitive advantage to the bacterium in colonizing the inflamed intestine of wild-type but not of lipocalin-2-deficient mice. Thus, resistance to lipocalin-2 defines a specific adaptation of S. Typhimurium for growth in the inflamed intestine.


Subject(s)
Host-Pathogen Interactions , Intestines/immunology , Lipocalins/immunology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Animals , Cell Line , Disease Models, Animal , Gene Expression , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukins/genetics , Interleukins/immunology , Intestines/microbiology , Lipocalins/genetics , Macaca mulatta , Mice , Mice, Inbred C57BL , Salmonella Infections/genetics , Salmonella typhimurium/immunology , Siderophores/immunology , Interleukin-22
12.
Protein Sci ; 18(5): 1081-94, 2009 May.
Article in English | MEDLINE | ID: mdl-19388054

ABSTRACT

Prostasin or human channel-activating protease 1 has been reported to play a critical role in the regulation of extracellular sodium ion transport via its activation of the epithelial cell sodium channel. Here, the structure of the extracellular portion of the membrane associated serine protease has been solved to high resolution in complex with a nonselective d-FFR chloromethyl ketone inhibitor, in an apo form, in a form where the apo crystal has been soaked with the covalent inhibitor camostat and in complex with the protein inhibitor aprotinin. It was also crystallized in the presence of the divalent cation Ca(+2). Comparison of the structures with each other and with other members of the trypsin-like serine protease family reveals unique structural features of prostasin and a large degree of conformational variation within specificity determining loops. Of particular interest is the S1 subsite loop which opens and closes in response to basic residues or divalent ions, directly binding Ca(+2) cations. This induced fit active site provides a new possible mode of regulation of trypsin-like proteases adapted in particular to extracellular regions with variable ionic concentrations such as the outer membrane layer of the epithelial cell.


Subject(s)
Catalytic Domain , Cations, Divalent/metabolism , Serine Endopeptidases/chemistry , Aprotinin/metabolism , Calcium/metabolism , Chymotrypsin/metabolism , Crystallography, X-Ray , Esters , Gabexate/analogs & derivatives , Gabexate/metabolism , Guanidines , Humans , Protease Inhibitors/metabolism , Protein Conformation , Sequence Alignment , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Substrate Specificity
13.
Gastroenterology ; 134(4): 1049-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395086

ABSTRACT

BACKGROUND & AIMS: We used the rhesus macaque model to study the effects of the cag pathogenicity island (cag PAI) on the H pylori host-pathogen interaction. METHODS: H pylori-specific pathogen-free (SPF) monkeys were experimentally challenged with wild-type (WT) H pylori strain J166 (J166WT, n = 4) or its cag PAI isogenic knockout (J166Deltacag PAI, n = 4). Animals underwent endoscopy before and 1, 4, 8, and 13 weeks after challenge. Gastric biopsies were collected for quantitative culture, histopathology, and host gene expression analysis. RESULTS: Quantitative cultures showed that all experimentally challenged animals were infected with J166WT or its isogenic J166Deltacag PAI. Histopathology demonstrated that inflammation and expansion of the lamina propria were attenuated in animals infected with J166Deltacag PAI compared with J166WT. Microarray analysis showed that of the 119 up-regulated genes in the J166WT-infected animals, several encode innate antimicrobial effector proteins, including elafin, siderocalin, DMBT1, DUOX2, and several novel paralogues of human-beta defensin-2. Quantitative RT-PCR confirmed that high-level induction of each of these genes depended on the presence of the cag PAI. Immunohistochemistry confirmed increased human-beta defensin-2 epithelial cell staining in animals challenged with J166WT compared with either J166Deltacag PAI-challenged or uninfected control animals. CONCLUSIONS: We propose that one function of the cag PAI is to induce an antimicrobial host response that may serve to increase the competitive advantage of H pylori in the gastric niche and could even provide a protective benefit to the host.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter pylori/pathogenicity , RNA, Bacterial/genetics , Animals , Biopsy , Calcium-Binding Proteins , Colony Count, Microbial , DNA-Binding Proteins , Disease Models, Animal , Dual Oxidases , Elafin/genetics , Elafin/metabolism , Endoscopy, Gastrointestinal , Female , Flavoproteins/genetics , Flavoproteins/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/metabolism , Gastritis/pathology , Gene Expression Regulation , Helicobacter pylori/growth & development , Helicobacter pylori/isolation & purification , Humans , Immunohistochemistry , Macaca mulatta , Male , Molecular Sequence Data , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins , beta-Defensins/genetics , beta-Defensins/metabolism
14.
J Mol Biol ; 373(5): 1243-54, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17900615

ABSTRACT

Pseudouridine (Psi) synthases catalyze the formation of one or more specific Psis in structured RNAs. Five families of Psi synthases have been characterized based on sequence homology. Pus10 has no significant sequence homology to these defined families and therefore represents a new family of Psi synthases. Initial characterization studies show that an archael Pus10 catalyzes the universally conserved Psi55 in tRNA. We present here the crystal structure of human Pus10 at 2.0 A resolution, which is the first structural description from this novel Psi synthase family. Pus10 is a crescent-shaped molecule with two domains, the universally conserved Psi synthase catalytic domain and a THUMP-containing domain, which is unique to the Pus10 family. Superposition of the catalytic domains of Pus10 and other Psi synthases identifies the full set of conserved Psi synthase active site residues indicating that Pus10 likely employs a similar catalytic mechanism to other Psi synthases. The Pus10 active site is located in a deep pocket of a basic cleft adjacent to flexible thumb and forefinger loops, which could provide further stabilization for binding the RNA substrate. Modeling studies demonstrate that the cleft between the catalytic and accessory domain is large enough and electrostatically compatible to accommodate an RNA stem and support the role of the N-terminal domain as an accessory RNA-binding domain.


Subject(s)
Hydro-Lyases/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , RNA/metabolism
15.
J Med Chem ; 50(11): 2693-9, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17469812

ABSTRACT

The substrate activity screening (SAS) method, a substrate-based fragment identification and optimization method for the development of enzyme inhibitors, was previously applied to cathepsin S to obtain a novel (2-arylphenoxy)acetaldehyde inhibitor, 2, with a 0.49 microM Ki value (Wood, W. J. L.; Patterson, A. W.; Tsuruoka, H.; Jain, R. K.; Ellman, J. A. J. Am. Chem. Soc. 2005, 127, 15521-15527). In this paper we disclose the X-ray structure of a complex between cathepsin S and inhibitor 2 which reveals an unprecedented binding mode. On the basis of this structure, additional 2-biaryloxy substrates with greatly increased cleavage efficiency were designed. Conversion of the optimized substrates to the corresponding aldehyde inhibitors yielded a low molecular weight (304 Daltons) and potent (9.6 nM) cathepsin S inhibitor that showed from 100- to >1000-fold selectivity relative to cathepsins B, L, and K.


Subject(s)
Acetaldehyde/analogs & derivatives , Acetaldehyde/chemical synthesis , Biphenyl Compounds/chemical synthesis , Cathepsins/antagonists & inhibitors , Cathepsins/chemistry , Models, Molecular , Acetaldehyde/chemistry , Biphenyl Compounds/chemistry , Crystallography, X-Ray , Protein Binding , Structure-Activity Relationship
16.
J Med Chem ; 49(21): 6298-307, 2006 Oct 19.
Article in English | MEDLINE | ID: mdl-17034136

ABSTRACT

The substrate activity screening method, a substrate-based fragment identification and optimization method for the development of enzyme inhibitors, was previously applied to cathepsin S to obtain low nanomolar 1,4-disubstituted-1,2,3-triazole-based aldehyde inhibitors (Wood, W. J. L.; Patterson, A. W.; Tsuruoka, H.; Jain, R. K.; Ellman, J. A. J. Am. Chem. Soc. 2005, 127, 15521-15527). Replacement of the metabolically labile aldehyde pharmacophore with the nitrile pharmacophore provided inhibitors with moderate potency for cathepsin S. The inhibitors showed good selectivity over cathepsins B and L but no selectivity over cathepsin K. X-ray structures of two crystal forms (1.5 and 1.9 A) of a complex between cathepsin S and a triazole inhibitor incorporating a chloromethyl ketone pharmacophore guided the design of triazole substrates with increased cleavage efficiency and selectivity for cathepsin S over cathepsins B, L, and K. Conversion of select substrates to nitrile inhibitors yielded a low molecular weight (414 Da) and potent (15 nM) cathepsin S inhibitor that showed >1000-fold selectivity over cathepsins B, L, and K.


Subject(s)
Cathepsins/antagonists & inhibitors , Cathepsins/chemistry , Nitriles/chemical synthesis , Protease Inhibitors/chemical synthesis , Triazoles/chemical synthesis , Amino Acid Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Molecular Structure , Nitriles/chemistry , Protease Inhibitors/chemistry , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity , Triazoles/chemistry
17.
Bioorg Med Chem Lett ; 16(19): 5112-7, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16876402

ABSTRACT

The synthesis and structure-activity relationship of a series of arylaminoethyl amide cathepsin S inhibitors are reported. Optimization of P3 and P2 groups to improve overall physicochemical properties resulted in significant improvements in oral bioavailability over early lead compounds. An X-ray structure of compound 37 bound to the active site of cathepsin S is also reported.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Cathepsins/antagonists & inhibitors , Administration, Oral , Amides/pharmacokinetics , Animals , Binding Sites , Biological Availability , Crystallography, X-Ray , Ethers, Cyclic/chemical synthesis , Ethers, Cyclic/pharmacology , Humans , Male , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Zinc
19.
Proteins ; 63(4): 1112-8, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16544291
SELECTION OF CITATIONS
SEARCH DETAIL
...