Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 142(4): 637-46, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20723762

ABSTRACT

Apoptosis is a conserved cellular pathway that results in the activation of cysteine-aspartyl proteases, or caspases. To dissect the nonredundant roles of the executioner caspase-3, -6, and -7 in orchestrating apoptosis, we have developed an orthogonal protease to selectively activate each isoform in human cells. Our approach uses a split-tobacco etch virus (TEV) protease under small-molecule control, which we call the SNIPer, with caspase alleles containing genetically encoded TEV cleavage sites. These studies reveal that all three caspases are transiently activated but only activation of caspase-3 or -7 is sufficient to induce apoptosis. Proteomic analysis shown here and from others reveals that 20 of the 33 subunits of the 26S proteasome can be cut by caspases, and we demonstrate synergy between proteasome inhibition and dose-dependent caspase activation. We propose a model of proteolytic reciprocal negative regulation with mechanistic implications for the combined clinical use of proteasome inhibitors and proapoptotic drugs.


Subject(s)
Apoptosis , Caspase 3/metabolism , Caspase 7/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Protein Engineering , Caspase 6/metabolism , Cell Line , Drug Design , Enzyme Activation/drug effects , Humans , Isoenzymes/metabolism , Leupeptins/pharmacology , Proteasome Endopeptidase Complex , Proteasome Inhibitors
2.
Cell ; 134(5): 866-76, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18722006

ABSTRACT

The nearly 600 proteases in the human genome regulate a diversity of biological processes, including programmed cell death. Comprehensive characterization of protease signaling in complex biological samples is limited by available proteomic methods. We have developed a general approach for global identification of proteolytic cleavage sites using an engineered enzyme to selectively biotinylate free protein N termini for positive enrichment of corresponding N-terminal peptides. Using this method to study apoptosis, we have sequenced 333 caspase-like cleavage sites distributed among 292 protein substrates. These sites are generally not predicted by in vitro caspase substrate specificity but can be used to predict other physiological caspase cleavage sites. Structural bioinformatic studies show that caspase cleavage sites often appear in surface-accessible loops and even occasionally in helical regions. Strikingly, we also find that a disproportionate number of caspase substrates physically interact, suggesting that these dimeric proteases target protein complexes and networks to elicit apoptosis.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Proteins/analysis , Proteins/metabolism , Proteomics , Antineoplastic Agents, Phytogenic/pharmacology , Caspases/chemistry , Etoposide/pharmacology , Humans , Jurkat Cells , Protein Binding , Substrate Specificity
3.
Mol Cell Proteomics ; 12(3): 813-24, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23264352

ABSTRACT

Proteolysis is a critical post-translational modification for regulation of cellular processes. Our lab has previously developed a technique for specifically labeling unmodified protein N termini, the α-aminome, using the engineered enzyme, subtiligase. Here we present a database, called the DegraBase (http://wellslab.ucsf.edu/degrabase/), which compiles 8090 unique N termini from 3206 proteins directly identified in subtiligase-based positive enrichment mass spectrometry experiments in healthy and apoptotic human cell lines. We include both previously published and unpublished data in our analysis, resulting in a total of 2144 unique α-amines identified in healthy cells, and 6990 in cells undergoing apoptosis. The N termini derive from three general categories of proteolysis with respect to cleavage location and functional role: translational N-terminal methionine processing (∼10% of total proteolysis), sites close to the translational N terminus that likely represent removal of transit or signal peptides (∼25% of total), and finally, other endoproteolytic cuts (∼65% of total). Induction of apoptosis causes relatively little change in the first two proteolytic categories, but dramatic changes are seen in endoproteolysis. For example, we observed 1706 putative apoptotic caspase cuts, more than double the total annotated sites in the CASBAH and MEROPS databases. In the endoproteolysis category, there are a total of nearly 3000 noncaspase nontryptic cleavages that are not currently reported in the MEROPS database. These studies significantly increase the annotation for all categories of proteolysis in human cells and allow public access for investigators to explore interesting proteolytic events in healthy and apoptotic human cells.


Subject(s)
Apoptosis , Databases, Protein , Proteolysis , Proteome/analysis , Proteomics/methods , Tandem Mass Spectrometry/methods , Caspases/metabolism , Cell Line, Tumor , Chromatography, Liquid/methods , Humans , Internet , Jurkat Cells , Peptide Synthases/metabolism , Peptides/analysis , Peptides/chemistry , Peptides/metabolism , Proteome/chemistry , Proteome/metabolism , Subtilisins/metabolism
4.
Proc Natl Acad Sci U S A ; 109(6): 1913-8, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308409

ABSTRACT

Mass spectrometry-based proteomics is a powerful tool for identifying hundreds to thousands of posttranslational modifications in complex mixtures. However, it remains enormously challenging to simultaneously assess the intrinsic catalytic efficiencies (k(cat)/K(M)) of these modifications in the context of their natural interactors. Such fundamental enzymological constants are key to determining substrate specificity and for establishing the timing and importance of cellular signaling. Here, we report the use of selected reaction monitoring (SRM) for tracking proteolysis induced by human apoptotic caspases-3, -7, -8, and -9 in lysates and living cells. By following the appearance of the cleaved peptides in lysate as a function of time, we were able to determine hundreds of catalytic efficiencies in parallel. Remarkably, we find the rates of substrate hydrolysis for individual caspases vary greater than 500-fold indicating a sequential process. Moreover, the rank-order of substrate cutting is similar in apoptotic cells, suggesting that cellular structures do not dramatically alter substrate accessibility. Comparisons of extrinsic (TRAIL) and intrinsic (staurosporine) inducers of apoptosis revealed similar substrate profiles, suggesting the final proteolytic demolitions proceed by similarly ordered plans. Certain biological processes were rapidly targeted by the caspases, including multiple components of the endocyotic pathway and miRNA processing machinery. We believe this massively parallel and quantitative label-free approach to obtaining basic enzymological constants will facilitate the study of proteolysis and other posttranslational modifications in complex mixtures.


Subject(s)
Proteolysis , Proteomics/methods , Amino Acid Sequence , Apoptosis/drug effects , Caspases/metabolism , Cell Survival/drug effects , Endocytosis/drug effects , Enzyme Activation/drug effects , Humans , Jurkat Cells , Kinetics , MicroRNAs/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Proteolysis/drug effects , RNA Processing, Post-Transcriptional/drug effects , Staurosporine/pharmacology , Substrate Specificity/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology
5.
Proc Natl Acad Sci U S A ; 109(31): 12432-7, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802652

ABSTRACT

Proapoptotic drugs are a mainstay of cancer drug treatment. These drugs stress cells and ultimately trigger the activation of caspases, cysteine-class proteases that cleave after aspartic acid and deconstruct the cell. It is well known that cells respond differently to proapoptotic cancer drug treatments. Here, using a global and unbiased quantitative N-terminomics technology, we show that ~500 products of caspase cleavage and their kinetics vary dramatically between cell type and cytotoxic drug treatment. It is likely that variations arise from differences in baseline proteome composition of the cell type and the alterations induced by drug treatments to yield a unique cohort of proteins that caspases finally target. Many targets are specific to both drug treatment and cell type, providing candidate-specific biomarkers for apoptosis. For example, in multiple myeloma cells treated with the proteasome inhibitor bortezomib, levels of activating transcription factor-4 increase dramatically early in drug treatment and then decrease upon cleavage by activated caspases. Thus, caspase-derived cleavage products are a sensitive reflection of cell-type and drug-induced stress, and provide useful fingerprints for mechanisms of drug action and response.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Caspases/metabolism , Cytotoxins/pharmacology , Multiple Myeloma/drug therapy , Neoplasm Proteins/metabolism , Activating Transcription Factor 4/metabolism , Antineoplastic Agents/chemistry , Boronic Acids/pharmacology , Bortezomib , Cytotoxins/chemistry , Drug Screening Assays, Antitumor/methods , Humans , Jurkat Cells , Kinetics , Multiple Myeloma/enzymology , Proteome/metabolism , Pyrazines/pharmacology
6.
Clin Cancer Res ; 28(13): 2844-2853, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35511917

ABSTRACT

PURPOSE: The utility of real-world data (RWD) for use as external controls in drug development is informed by studies that replicate trial control arms for different endpoints. The purpose of this study was to replicate control arms from four non-small cell lung cancer (NSCLC) randomized controlled trials (RCT) to analyze overall survival (OS), progression-free survival (PFS), and overall response rate (ORR) using RWD. PATIENTS AND METHODS: This study used RWD from a nationwide de-identified database and a clinico-genomic database to replicate OS, PFS, and ORR endpoints in the chemotherapy control arms of four first-line NSCLC RCTs evaluating atezolizumab [IMpower150-wild-type (WT), IMpower130-WT, IMpower131, and IMpower132]. Additional objectives were to develop a definition of real-world PFS (rwPFS) and to evaluate the real-world response rate (rwRR) endpoint. RESULTS: Baseline demographic and clinical characteristics were balanced after application of propensity score weighting methods. For rwPFS and OS, RWD external controls were generally similar to their RCT control counterparts. Across all four trials, the hazard ratio (HR) point estimates comparing trial controls with external controls were closer to 1.0 for the PFS endpoint than for the OS endpoint. An exploratory assessment of rwRR in RWD revealed a slight but nonsignificant overestimation of RCT ORR, which was unconfounded by baseline characteristics. CONCLUSIONS: RWD can be used to reasonably replicate the OS and PFS of chemotherapy control arms of first-line NSCLC RCTs. Additional studies can provide greater insight into the utility of RWD in drug development.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Progression-Free Survival , Proportional Hazards Models , Randomized Controlled Trials as Topic
7.
Bioinformatics ; 26(14): 1714-22, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20505003

ABSTRACT

MOTIVATION: Granzyme B (GrB) and caspases cleave specific protein substrates to induce apoptosis in virally infected and neoplastic cells. While substrates for both types of proteases have been determined experimentally, there are many more yet to be discovered in humans and other metazoans. Here, we present a bioinformatics method based on support vector machine (SVM) learning that identifies sequence and structural features important for protease recognition of substrate peptides and then uses these features to predict novel substrates. Our approach can act as a convenient hypothesis generator, guiding future experiments by high-confidence identification of peptide-protein partners. RESULTS: The method is benchmarked on the known substrates of both protease types, including our literature-curated GrB substrate set (GrBah). On these benchmark sets, the method outperforms a number of other methods that consider sequence only, predicting at a 0.87 true positive rate (TPR) and a 0.13 false positive rate (FPR) for caspase substrates, and a 0.79 TPR and a 0.21 FPR for GrB substrates. The method is then applied to approximately 25 000 proteins in the human proteome to generate a ranked list of predicted substrates of each protease type. Two of these predictions, AIF-1 and SMN1, were selected for further experimental analysis, and each was validated as a GrB substrate. AVAILABILITY: All predictions for both protease types are publically available at http://salilab.org/peptide. A web server is at the same site that allows a user to train new SVM models to make predictions for any protein that recognizes specific oligopeptide ligands.


Subject(s)
Computational Biology/methods , Peptide Hydrolases/chemistry , Sequence Analysis, Protein/methods , Caspases/chemistry , Ligands
8.
Clin Cancer Res ; 25(11): 3220-3228, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30770348

ABSTRACT

PURPOSE: IDO1 induces immune suppression in T cells through l-tryptophan (Trp) depletion and kynurenine (Kyn) accumulation in the local tumor microenvironment, suppressing effector T cells and hyperactivating regulatory T cells (Treg). Navoximod is an investigational small-molecule inhibitor of IDO1. This phase I study evaluated safety, tolerability, pharmacokinetics, and pharmacodynamics of navoximod in combination with atezolizumab, a PD-L1 inhibitor, in patients with advanced cancer. PATIENTS AND METHODS: The study consisted of a 3+3 dose-escalation stage (n = 66) and a tumor-specific expansion stage (n = 92). Navoximod was given orally every 12 hours continuously for 21 consecutive days of each cycle with the exception of cycle 1, where navoximod administration started on day -1 to characterize pharmacokinetics. Atezolizumab was administered by intravenous infusion 1,200 mg every 3 weeks on day 1 of each cycle. RESULTS: Patients (n = 157) received navoximod at 6 dose levels (50-1,000 mg) in combination with atezolizumab. The maximum administered dose was 1,000 mg twice daily; the MTD was not reached. Navoximod demonstrated a linear pharmacokinetic profile, and plasma Kyn generally decreased with increasing doses of navoximod. The most common treatment-related AEs were fatigue (22%), rash (22%), and chromaturia (20%). Activity was observed at all dose levels in various tumor types (melanoma, pancreatic, prostate, ovarian, head and neck squamous cell carcinoma, cervical, neural sheath, non-small cell lung cancer, triple-negative breast cancer, renal cell carcinoma, urothelial bladder cancer): 6 (9%) dose-escalation patients achieved partial response, and 10 (11%) expansion patients achieved partial response or complete response. CONCLUSIONS: The combination of navoximod and atezolizumab demonstrated acceptable safety, tolerability, and pharmacokinetics for patients with advanced cancer. Although activity was observed, there was no clear evidence of benefit from adding navoximod to atezolizumab.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacokinetics , Indoles/administration & dosage , Indoles/pharmacokinetics , Magnetic Resonance Imaging , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/etiology , Neoplasms/metabolism , Tomography, X-Ray Computed , Treatment Outcome
9.
Bioorg Med Chem Lett ; 18(22): 6000-3, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18762420

ABSTRACT

The peptide ligase subtiligase, derived from subtilisin, has been employed in the identification of protein N-termini in complex mixtures. Here, the peptide ester substrates for the ligation reaction were optimized with respect to solubility, resulting in greater incorporation of the N-terminal tags. Additionally, the quantitation of the incorporated tags was explored, and a 'click' chemistry-based derivatization provided the ability to quantitate the tag to low nanomolar concentrations by sandwich ELISA. These new tags should expand the utility of subtiligase for the proteomic study of N-termini.


Subject(s)
Isotope Labeling , Peptide Synthases/chemistry , Proteins/analysis , Proteomics , Subtilisins/chemistry , Alkynes/analysis , Alkynes/chemistry , Amino Acid Sequence , Enzyme-Linked Immunosorbent Assay , Glycine/analogs & derivatives , Glycine/analysis , Glycine/chemistry , Humans , Jurkat Cells , Proteins/chemistry , Solubility , Structure-Activity Relationship , Substrate Specificity , Tyrosine/analogs & derivatives , Tyrosine/analysis , Tyrosine/chemistry
10.
Bioanalysis ; 10(16): 1307-1317, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30141341

ABSTRACT

AIM: Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) catalyze the initial and rate-controlling step of tryptophan metabolism through the kynurenine pathway, which plays an important role in mediating immune response. Accurate measurement of tryptophan and kynurenine is critical for monitoring the activity of IDO/TDO. Experimental: Surrogate analytes ([15N2]-Tryptophan and [13C6]-Kynurenine) were used for preparation of calibration standard and quality control. A fit-for-purpose validation using an approach of surrogate analyte and authentic matrix was carried out. RESULTS: Acid precipitation was used in sample preparation, which yielded good recovery without significant matrix effect. Precision and accuracy results were well within the acceptance criteria. The assay demonstrated successful application to a clinical study to confirm a transient depletion of kynurenine upon IDO inhibition. CONCLUSION: A robust, specific and simple LC-MS/MS method was developed and validated with a fit-for-purpose style for measuring tryptophan and kynurenine in human plasma samples.


Subject(s)
Blood Chemical Analysis/methods , Kynurenine/blood , Tandem Mass Spectrometry , Tryptophan/blood , Biomarkers/blood , Biomarkers/metabolism , Chromatography, Liquid , Enzyme Inhibitors/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Limit of Detection , Tryptophan/metabolism
11.
J Immunother Cancer ; 6(1): 61, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29921320

ABSTRACT

BACKGROUND: Indoleamine-2,3-dioxygenase 1 (IDO1) catalyzes the oxidation of tryptophan into kynurenine and is partially responsible for acquired immune tolerance associated with cancer. The IDO1 small molecule inhibitor navoximod (GDC-0919, NLG-919) is active as a combination therapy in multiple tumor models. METHODS: This open-label Phase Ia study assessed safety, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary anti-tumor activity of navoximod in patients with recurrent/advanced solid tumors, administered as 50-800 mg BID on a 21/28 day and at 600 mg on a 28/28 day schedule. Plasma kynurenine and tryptophan were longitudinally evaluated and tumor assessments were performed. RESULTS: Patients (n = 22) received a median of 3 cycles of navoximod. No maximum tolerated dose was reached. One dose-limiting toxicity of Grade 4 lower gastrointestinal hemorrhage was reported. Adverse events (AEs) regardless of causality in ≥20% of patients included fatigue (59%), cough, decreased appetite, and pruritus (41% each), nausea (36%), and vomiting (27%). Grade ≥ 3 AEs occurred in 14/22 patients (64%), and were related to navoximod in two patients (9%). Navoximod was rapidly absorbed (Tmax ~ 1 h) and exhibited dose-proportional increases in exposure, with a half-life (t1/2 ~ 11 h) supportive of BID dosing. Navoximod transiently decreased plasma kynurenine from baseline levels with kinetics consistent with its half-life. Of efficacy-evaluable patients, 8 (36%) had stable disease and 10 (46%) had progressive disease. CONCLUSIONS: Navoximod was well-tolerated at doses up to 800 mg BID decreasing plasma kynurenine levels consistent with its half-life. Stable disease responses were observed. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02048709 .


Subject(s)
Enzyme Inhibitors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/enzymology , Recurrence , Treatment Outcome
12.
Clin Cancer Res ; 23(10): 2423-2432, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27815358

ABSTRACT

Purpose: Chk1 inhibition potentiates DNA-damaging chemotherapy by overriding cell-cycle arrest and genome repair. This phase I study evaluated the Chk1 inhibitor GDC-0425 given in combination with gemcitabine to patients with advanced solid tumors.Experimental Design: Patients received GDC-0425 alone for a 1-week lead-in followed by 21-day cycles of gemcitabine plus GDC-0425. Gemcitabine was initially administered at 750 mg/m2 (Arm A), then increased to 1,000 mg/m2 (Arm B), on days 1 and 8 in a 3 + 3 + 3 dose escalation to establish maximum tolerated dose (MTD). GDC-0425 was initially administered daily for three consecutive days; however, dosing was abbreviated to a single day on the basis of pharmacokinetics and tolerability. TP53 mutations were evaluated in archival tumor tissue. On-treatment tumor biopsies underwent pharmacodynamic biomarker analyses.Results: Forty patients were treated with GDC-0425. The MTD of GDC-0425 was 60 mg when administered approximately 24 hours after gemcitabine 1,000 mg/m2 Dose-limiting toxicities included thrombocytopenia (n = 5), neutropenia (n = 4), dyspnea, nausea, pyrexia, syncope, and increased alanine aminotransferase (n = 1 each). Common related adverse events were nausea (48%); anemia, neutropenia, vomiting (45% each); fatigue (43%); pyrexia (40%); and thrombocytopenia (35%). The GDC-0425 half-life was approximately 15 hours. There were two confirmed partial responses in patients with triple-negative breast cancer (TP53-mutated) and melanoma (n = 1 each) and one unconfirmed partial response in a patient with cancer of unknown primary origin.Conclusions: Chk1 inhibition with GDC-0425 in combination with gemcitabine was tolerated with manageable bone marrow suppression. The observed preliminary clinical activity warrants further investigation of this chemopotentiation strategy. Clin Cancer Res; 23(10); 2423-32. ©2016 AACR.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/administration & dosage , Melanoma/drug therapy , Piperidines/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Checkpoint Kinase 1/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Heterocyclic Compounds, 3-Ring/adverse effects , Humans , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Piperidines/adverse effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Gemcitabine
13.
Chem Biol ; 12(5): 567-77, 2005 May.
Article in English | MEDLINE | ID: mdl-15911377

ABSTRACT

The mechanism of target cell lysis in cytotoxic lymphocyte-mediated death is not well understood, and the role of granzymes in this process is unclear. Chemical functional probes were thus prepared for the major granzymes A and B to deconvolute their role in natural killer cell-mediated lysis of target cells. These biotinylated and substrate specificity-based diphenyl phosphonates allowed facile evaluation of selectivity through activity-based profiling in cell lysates and intact cells. Both inhibitors were found to be extremely selective in vitro and in cells. Use of these inhibitors in cell-based assays revealed granzyme A to be a minor effector and granzyme B to be a major effector of target cell lysis by natural killer cells. These studies indicate that the proapoptotic granzyme B functions also as a pronecrotic effector of target cell death.


Subject(s)
Killer Cells, Natural/enzymology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Apoptosis , Avidin/metabolism , Cell Death , Cell Line , Granzymes , Humans , K562 Cells , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology , Serine Proteinase Inhibitors/chemical synthesis
14.
J Am Chem Soc ; 128(32): 10362-3, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-16895390

ABSTRACT

Chemical cross-linking followed by identification of the cross-linked residues by mass spectrometry provides structural information on protein interaction surfaces. Nevertheless, accurate analysis of the digested, cross-linked proteins is often challenging. Herein, we describe a novel strategy that relies on the use of affinity-tagged cross-linkers and isotope coding on the cross-linker-modified species. Incorporation of O16 or O18 during the hydrolysis of the cross-linkers results in a characteristic "doublet" for the undesired products of a half-cross-linking reaction. Therefore, genuine cross-linked peptides are readily distinguished for further structural analysis. This strategy permits a sensitive and facile analysis on a dimeric protease inhibitor, ecotin, showing general applicability to other protein assemblies.


Subject(s)
Affinity Labels , Cross-Linking Reagents/chemistry , Isotope Labeling/methods , Proteins/chemistry , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Models, Molecular , Molecular Structure , Periplasmic Proteins/chemistry , Surface Properties
15.
J Biol Chem ; 279(52): 54275-82, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15494398

ABSTRACT

Granzyme M is a trypsin-fold serine protease that is specifically found in the granules of natural killer cells. This enzyme has been implicated recently in the induction of target cell death by cytotoxic lymphocytes, but unlike granzymes A and B, the molecular mechanism of action of granzyme M is unknown. We have characterized the extended substrate specificity of human granzyme M by using purified recombinant enzyme, several positional scanning libraries of coumarin substrates, and a panel of individual p-nitroanilide and coumarin substrates. In contrast to previous studies conducted using thiobenzyl ester substrates (Smyth, M. J., O'Connor, M. D., Trapani, J. A., Kershaw, M. H., and Brinkworth, R. I. (1996) J. Immunol. 156, 4174-4181), a strong preference for leucine at P1 over methionine was demonstrated. The extended substrate specificity was determined to be lysine = norleucine at P4, broad at P3, proline > alanine at P2, and leucine > norleucine > methionine at P1. The enzyme activity was found to be highly dependent on the length and sequence of substrates, indicative of a regulatory function for human granzyme M. Finally, the interaction between granzyme M and the serpins alpha(1)-antichymotrypsin, alpha(1)-proteinase inhibitor, and proteinase inhibitor 9 was characterized by using a candidate-based approach to identify potential endogenous inhibitors. Proteinase inhibitor 9 was effectively hydrolyzed and inactivated by human granzyme M, raising the possibility that this orphan granzyme bypasses proteinase inhibitor 9 inhibition of granzyme B.


Subject(s)
Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Serpins/metabolism , Serpins/pharmacology , Amino Acid Sequence , Binding Sites , Coumarins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/metabolism , Granzymes , Humans , Hydrolysis , Leucine/metabolism , Molecular Sequence Data , Norleucine/metabolism , Peptide Library , Proline/metabolism , Recombinant Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship , Substrate Specificity , alpha 1-Antichymotrypsin/metabolism , alpha 1-Antitrypsin/metabolism
16.
Nat Struct Biol ; 10(7): 527-34, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12819769

ABSTRACT

The cell death-inducing serine protease granzyme A (GzmA) has a unique disulfide-linked quaternary structure. The structure of human GzmA bound to a tripeptide CMK inhibitor, determined at a resolution of 2.4 A, reveals that the oligomeric state contributes to substrate selection by limiting access to the active site for potential macromolecular substrates and inhibitors. Unlike other serine proteases, tetrapeptide substrate preferences do not correlate well with natural substrate cleavage sequences. This suggests that the context of the cleavage sequence within a macromolecular substrate imposes another level of selection not observed with the peptide substrates. Modeling of inhibitors bound to the GzmA active site shows that the dimer also contributes to substrate specificity in a unique manner by extending the active-site cleft. The crystal structure, along with substrate library profiling and mutagenesis, has allowed us to identify and rationally manipulate key components involved in GzmA substrate specificity.


Subject(s)
Biopolymers/chemistry , Serine Endopeptidases/chemistry , Amino Acid Sequence , Biopolymers/metabolism , Dimerization , Granzymes , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Serine Endopeptidases/metabolism , Substrate Specificity
17.
J Virol ; 76(3): 1359-68, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11773410

ABSTRACT

Resistance to human immunodeficiency virus type 1 protease (HIV PR) inhibitors results primarily from the selection of multiple mutations in the protease region. Because many of these mutations are selected for the ability to decrease inhibitor binding in the active site, they also affect substrate binding and potentially substrate specificity. This work investigates the substrate specificity of a panel of clinically derived protease inhibitor-resistant HIV PR variants. To compare protease specificity, we have used positional-scanning, synthetic combinatorial peptide libraries as well as a select number of individual substrates. The subsite preferences of wild-type HIV PR determined by using the substrate libraries are consistent with prior reports, validating the use of these libraries to compare specificity among a panel of HIV PR variants. Five out of seven protease variants demonstrated subtle differences in specificity that may have significant impacts on their abilities to function in viral maturation. Of these, four variants demonstrated up to fourfold changes in the preference for valine relative to alanine at position P2 when tested on individual peptide substrates. This change correlated with a common mutation in the viral NC/p1 cleavage site. These mutations may represent a mechanism by which severely compromised, drug-resistant viral strains can increase fitness levels. Understanding the altered substrate specificity of drug-resistant HIV PR should be valuable in the design of future generations of protease inhibitors as well as in elucidating the molecular basis of regulation of proteolysis in HIV.


Subject(s)
Drug Resistance, Viral/genetics , HIV Protease/genetics , HIV-1/enzymology , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Catalytic Domain , Gene Products, gag/metabolism , HIV Protease/drug effects , HIV Protease/metabolism , HIV-1/drug effects , HIV-1/genetics , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Substrate Specificity , Valine/genetics , Valine/metabolism , gag Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL