Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(1): e0190255, 2018.
Article in English | MEDLINE | ID: mdl-29364904

ABSTRACT

Protein palmitoylation is a dynamic post-translational modification (PTM) important for cellular functions such as protein stability, trafficking, localization, and protein-protein interactions. S-palmitoylation occurs via the addition of palmitate to cysteine residues via a thioester linkage, catalyzed by palmitoyl acyl transferases (PATs), with removal of the palmitate catalyzed by acyl protein thioesterases (APTs) and palmitoyl-protein thioesterases (PPTs). Tools that target the regulators of palmitoylation-PATs, APTs and PPTs-will improve understanding of this essential PTM. Here, we describe the synthesis and application of a cell-permeable activity-based probe (ABP) that targets APTs in intact mammalian cells and the parasite Toxoplasma gondii. Using a focused library of substituted chloroisocoumarins, we identified a probe scaffold with nanomolar affinity for human APTs (HsAPT1 and HsAPT2) and synthesized a fluorescent ABP, JCP174-BODIPY TMR (JCP174-BT). We use JCP174-BT to profile HsAPT activity in situ in mammalian cells, to detect an APT in T. gondii (TgPPT1). We show discordance between HsAPT activity levels and total protein concentration in some cell lines, indicating that total protein levels may not be representative of APT activity in complex systems, highlighting the utility of this probe.


Subject(s)
Molecular Probes/metabolism , Animals , Mammals , Protein Processing, Post-Translational , Thiolester Hydrolases , Toxoplasma/enzymology
2.
PLoS Pathog ; 13(9): e1006586, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28922424

ABSTRACT

Proteases have been implicated in a variety of developmental processes during the malaria parasite lifecycle. In particular, invasion and egress of the parasite from the infected hepatocyte and erythrocyte, critically depend on protease activity. Although falcipain-1 was the first cysteine protease to be characterized in P. falciparum, its role in the lifecycle of the parasite has been the subject of some controversy. While an inhibitor of falcipain-1 blocked erythrocyte invasion by merozoites, two independent studies showed that falcipain-1 disruption did not affect growth of blood stage parasites. To shed light on the role of this protease over the entire Plasmodium lifecycle, we disrupted berghepain-1, its ortholog in the rodent parasite P. berghei. We found that this mutant parasite displays a pronounced delay in blood stage infection after inoculation of sporozoites. Experiments designed to pinpoint the defect of berghepain-1 knockout parasites found that it was not due to alterations in gliding motility, hepatocyte invasion or liver stage development and that injection of berghepain-1 knockout merosomes replicated the phenotype of delayed blood stage growth after sporozoite inoculation. We identified an additional role for berghepain-1 in preparing blood stage merozoites for infection of erythrocytes and observed that berghepain-1 knockout parasites exhibit a reticulocyte restriction, suggesting that berghepain-1 activity broadens the erythrocyte repertoire of the parasite. The lack of berghepain-1 expression resulted in a greater reduction in erythrocyte infectivity in hepatocyte-derived merozoites than it did in erythrocyte-derived merozoites. These observations indicate a role for berghepain-1 in processing ligands important for merozoite infectivity and provide evidence supporting the notion that hepatic and erythrocytic merozoites, though structurally similar, are not identical.


Subject(s)
Cysteine Endopeptidases/metabolism , Hepatocytes/metabolism , Malaria/metabolism , Merozoites/metabolism , Plasmodium falciparum/metabolism , Animals , Cysteine Proteinase Inhibitors/pharmacology , Erythrocytes/parasitology , Hepatocytes/parasitology , Liver/metabolism , Malaria/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism
3.
mBio ; 8(1)2017 02 28.
Article in English | MEDLINE | ID: mdl-28246362

ABSTRACT

Human DJ-1 is a highly conserved and yet functionally enigmatic protein associated with a heritable form of Parkinson's disease. It has been suggested to be a redox-dependent regulatory scaffold, binding to proteins to modulate their function. Here we present the X-ray crystal structure of the Toxoplasma orthologue Toxoplasma gondii DJ-1 (TgDJ-1) at 2.1-Å resolution and show that it directly associates with calcium-dependent protein kinase 1 (CDPK1). The TgDJ-1 structure identifies an orthologously conserved arginine dyad that acts as a phospho-gatekeeper motif to control complex formation. We determined that the binding of TgDJ-1 to CDPK1 is sensitive to oxidation and calcium, and that this interaction potentiates CDPK1 kinase activity. Finally, we show that genetic deletion of TgDJ-1 results in upregulation of CDPK1 expression and that disruption of the CDPK1/TgDJ-1 complex in vivo prevents normal exocytosis of parasite virulence-associated organelles called micronemes. Overall, our data suggest that TgDJ-1 functions as a noncanonical kinase-regulatory scaffold that integrates multiple intracellular signals to tune microneme exocytosis in T. gondiiIMPORTANCE Apicomplexan parasites such as Toxoplasma and Plasmodium are obligate intracellular parasites that require the protective environment of a host cell in order to replicate and survive within a host organism. These parasites secrete effector proteins from specialized apical organelles to select and invade a chosen host cell. The secretion of these organelles is a tightly regulated process coordinated by endogenous small molecules and calcium-dependent protein kinases. We previously identified the Toxoplasma orthologue of the highly conserved protein DJ-1 as a regulator of microneme secretion, but the molecular basis for this was not known. We have now identified the molecular mechanism for how TgDJ-1 regulates microneme secretion. TgDJ-1 interacts with the kinase responsible for the secretion of these organelles (calcium-dependent kinase 1) and synergizes with calcium to potentiate kinase activity. This interaction is direct, phosphodependent, and necessary for the normal secretion of these important organelles.


Subject(s)
Exosomes/metabolism , Protein Deglycase DJ-1/chemistry , Protein Deglycase DJ-1/metabolism , Protein Kinases/metabolism , Toxoplasma/enzymology , Toxoplasma/metabolism , Calcium/metabolism , Crystallography, X-Ray , Exocytosis , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Conformation
4.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G548-60, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27514475

ABSTRACT

Pancreatitis is an inflammatory disease of the pancreas characterized by dysregulated activity of digestive enzymes, necrosis, immune infiltration, and pain. Repeated incidence of pancreatitis is an important risk factor for pancreatic cancer. Legumain, a lysosomal cysteine protease, has been linked to inflammatory diseases such as atherosclerosis, stroke, and cancer. Until now, legumain activation has not been studied during pancreatitis. We used a fluorescently quenched activity-based probe to assess legumain activation during caerulein-induced pancreatitis in mice. We detected activated legumain by ex vivo imaging, confocal microscopy, and gel electrophoresis. Compared with healthy controls, legumain activity in the pancreas of caerulein-treated mice was increased in a time-dependent manner. Legumain was localized to CD68(+) macrophages and was not active in pancreatic acinar cells. Using a small-molecule inhibitor of legumain, we found that this protease is not essential for the initiation of pancreatitis. However, it may serve as a biomarker of disease, since patients with chronic pancreatitis show strongly increased legumain expression in macrophages. Moreover, the occurrence of legumain-expressing macrophages in regions of acinar-to-ductal metaplasia suggests that this protease may influence reprogramming events that lead to inflammation-induced pancreatic cancer.


Subject(s)
Cysteine Endopeptidases/metabolism , Macrophages/enzymology , Pancreatitis/enzymology , Animals , Ceruletide/toxicity , Cysteine Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced
5.
Cell Chem Biol ; 23(7): 793-804, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27427229

ABSTRACT

Cysteine cathepsins are lysosomal proteases involved in regulation of both normal cellular processes and disease. Biochemical studies with peptide substrates indicate that cathepsins have optimal activity at acidic pH and highly attenuated activity at neutral pH. In contrast, there is mounting evidence that cathepsins have biological roles in environments that have non-acidic pH. To further define the specific pH environments where cathepsins act, we designed bifunctional activity-based probes (ABPs) that allow simultaneous analysis of cathepsin protease activity and pH. We use these probes to analyze the steady-state environment of cathepsin activity in macrophages and to measure dynamic changes in activity and pH upon stimulation. We show that Salmonella typhimurium induces a change in lysosomal pH that ultimately impairs cathepsin activity in both infected cells and a fraction of bystander cells, highlighting a mechanism by which Salmonella can simultaneously flourish within host cells and alter the behavior of nearby uninfected cells.


Subject(s)
Bacterial Infections/metabolism , Cathepsins/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Molecular Probes/metabolism , Salmonella typhimurium/metabolism , Animals , Cathepsins/chemistry , Endosomes/chemistry , Hydrogen-Ion Concentration , Lysosomes/chemistry , Mice , Molecular Probes/chemistry , RAW 264.7 Cells , Salmonella typhimurium/growth & development , Salmonella typhimurium/isolation & purification
6.
ACS Infect Dis ; 2(3): 173-179, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-27347558

ABSTRACT

The recent Ebola virus outbreak in western Africa highlights the need for novel therapeutics that target Ebola virus and other filoviruses. Filoviruses require processing by host cell-derived cysteine cathepsins for productive infection. Here we report the generation of a focused library of cysteine cathepsin inhibitors and subsequent screening to identify compounds with potent activity against viral entry and replication. Our top compounds show highly potent and broad-spectrum activity against cysteine cathepsins and were able to effectively block entry of Ebola and Marburg viruses. These agents are promising leads for development as antifilovirus therapeutics.

7.
Elife ; 5: e13663, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27011353

ABSTRACT

When innate immune cells such as macrophages are challenged with environmental stresses or infection by pathogens, they trigger the rapid assembly of multi-protein complexes called inflammasomes that are responsible for initiating pro-inflammatory responses and a form of cell death termed pyroptosis. We describe here the identification of an intracellular trigger of NLRP3-mediated inflammatory signaling, IL-1ß production and pyroptosis in primed murine bone marrow-derived macrophages that is mediated by the disruption of glycolytic flux. This signal results from a drop of NADH levels and induction of mitochondrial ROS production and can be rescued by addition of products that restore NADH production. This signal is also important for host-cell response to the intracellular pathogen Salmonella typhimurium, which can disrupt metabolism by uptake of host-cell glucose. These results reveal an important inflammatory signaling network used by immune cells to sense metabolic dysfunction or infection by intracellular pathogens.


Subject(s)
Glycolysis , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Pyroptosis , Signal Transduction , Animals , Cells, Cultured , Interleukin-1beta/metabolism , Mice , NAD/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism
8.
Nature ; 530(7589): 233-6, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26863983

ABSTRACT

The proteasome is a multi-component protease complex responsible for regulating key processes such as the cell cycle and antigen presentation. Compounds that target the proteasome are potentially valuable tools for the treatment of pathogens that depend on proteasome function for survival and replication. In particular, proteasome inhibitors have been shown to be toxic for the malaria parasite Plasmodium falciparum at all stages of its life cycle. Most compounds that have been tested against the parasite also inhibit the mammalian proteasome, resulting in toxicity that precludes their use as therapeutic agents. Therefore, better definition of the substrate specificity and structural properties of the Plasmodium proteasome could enable the development of compounds with sufficient selectivity to allow their use as anti-malarial agents. To accomplish this goal, here we use a substrate profiling method to uncover differences in the specificities of the human and P. falciparum proteasome. We design inhibitors based on amino-acid preferences specific to the parasite proteasome, and find that they preferentially inhibit the ß2-subunit. We determine the structure of the P. falciparum 20S proteasome bound to the inhibitor using cryo-electron microscopy and single-particle analysis, to a resolution of 3.6 Å. These data reveal the unusually open P. falciparum ß2 active site and provide valuable information about active-site architecture that can be used to further refine inhibitor design. Furthermore, consistent with the recent finding that the proteasome is important for stress pathways associated with resistance of artemisinin family anti-malarials, we observe growth inhibition synergism with low doses of this ß2-selective inhibitor in artemisinin-sensitive and -resistant parasites. Finally, we demonstrate that a parasite-selective inhibitor could be used to attenuate parasite growth in vivo without appreciable toxicity to the host. Thus, the Plasmodium proteasome is a chemically tractable target that could be exploited by next-generation anti-malarial agents.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Drug Design , Plasmodium/drug effects , Plasmodium/enzymology , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacology , Animals , Antimalarials/adverse effects , Antimalarials/toxicity , Artemisinins/pharmacology , Catalytic Domain , Cryoelectron Microscopy , Dose-Response Relationship, Drug , Drug Resistance , Drug Synergism , Enzyme Activation , Female , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Plasmodium/growth & development , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/enzymology , Plasmodium chabaudi/physiology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/ultrastructure , Proteasome Inhibitors/adverse effects , Proteasome Inhibitors/toxicity , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Species Specificity , Substrate Specificity/drug effects
9.
Sci Rep ; 6: 19755, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26797565

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a lethal, chronic, progressive disease characterized by formation of scar tissue within the lungs. Because it is a disease of unknown etiology, it is difficult to diagnose, to predict disease course and to devise treatment strategies. Recent evidence suggests that activated macrophages play key roles in the pathology of IPF. Therefore, imaging probes that specifically recognize these pools of activated immune cells could provide valuable information about how these cells contribute to the pathobiology of the disease. Here we demonstrate that cysteine cathepsin-targeted imaging probes can be used to monitor the contribution of macrophages to fibrotic disease progression in the bleomycin-induced murine model of pulmonary fibrosis. Furthermore, we show that the probes highlight regions of macrophage involvement in fibrosis in human biopsy tissues from IPF patients. Finally, we present first-in-human results demonstrating non-invasive imaging of active cathepsins in fibrotic lesions of patients with IPF. Together, our findings validate small molecule cysteine cathepsin probes for clinical PET imaging and suggest that they have the potential to be used to generate mechanistically-informative molecular information regarding cellular drivers of IPF disease severity and progression.


Subject(s)
Cathepsins/metabolism , Diagnostic Imaging/methods , Idiopathic Pulmonary Fibrosis/diagnosis , Molecular Probes/metabolism , Animals , Bleomycin , Copper Radioisotopes , Disease Models, Animal , Disease Progression , Gallium Radioisotopes , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Macrophage Activation , Molecular Probes/chemistry , Optical Imaging , Positron-Emission Tomography
10.
Cell Host Microbe ; 18(4): 501-11, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468752

ABSTRACT

Post-translational modifications (PTMs) such as palmitoylation are critical for the lytic cycle of the protozoan parasite Toxoplasma gondii. While palmitoylation is involved in invasion, motility, and cell morphology, the proteins that utilize this PTM remain largely unknown. Using a chemical proteomic approach, we report a comprehensive analysis of palmitoylated proteins in T. gondii, identifying a total of 282 proteins, including cytosolic, membrane-associated, and transmembrane proteins. From this large set of palmitoylated targets, we validate palmitoylation of proteins involved in motility (myosin light chain 1, myosin A), cell morphology (PhIL1), and host cell invasion (apical membrane antigen 1, AMA1). Further studies reveal that blocking AMA1 palmitoylation enhances the release of AMA1 and other invasion-related proteins from apical secretory organelles, suggesting a previously unrecognized role for AMA1. These findings suggest that palmitoylation is ubiquitous throughout the T. gondii proteome and reveal insights into the biology of this important human pathogen.


Subject(s)
Palmitic Acids/metabolism , Protein Processing, Post-Translational , Proteins/analysis , Proteins/metabolism , Proteome/analysis , Toxoplasma/chemistry , Endocytosis , Humans , Locomotion , Toxoplasma/cytology , Toxoplasma/physiology , Virulence
11.
Oncotarget ; 6(29): 27008-22, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26308073

ABSTRACT

Cysteine cathepsin proteases contribute to many normal cellular functions, and their aberrant activity within various cell types can contribute to many diseases, including breast cancer. It is now well accepted that cathepsin proteases have numerous cell-specific functions within the tumor microenvironment that function to promote tumor growth and invasion, such that they may be valid targets for anti-metastatic therapeutic approaches. Using activity-based probes, we have examined the activity and expression of cysteine cathepsins in a mouse model of breast cancer metastasis to bone. In mice bearing highly metastatic tumors, we detected abundant cysteine cathepsin expression and activity in myeloid-derived suppressor cells (MDSCs). These immature immune cells have known metastasis-promoting roles, including immunosuppression and osteoclastogenesis, and we assessed the contribution of cysteine cathepsins to these functions. Blocking cysteine cathepsin activity with multiple small-molecule inhibitors resulted in enhanced differentiation of multinucleated osteoclasts. This highlights a potential role for cysteine cathepsin activity in suppressing the fusion of osteoclast precursor cells. In support of this hypothesis, we found that expression and activity of key cysteine cathepsins were downregulated during MDSC-osteoclast differentiation. Another cysteine protease, legumain, also inhibits osteoclastogenesis, in part through modulation of cathepsin L activity. Together, these data suggest that cysteine protease inhibition is associated with enhanced osteoclastogenesis, a process that has been implicated in bone metastasis.


Subject(s)
Cathepsins/metabolism , Cysteine/metabolism , Mammary Neoplasms, Animal/metabolism , Myeloid Cells/cytology , Osteoclasts/metabolism , Animals , Bone Neoplasms/secondary , Cathepsin L/chemistry , Cell Differentiation , Cell Separation , Cysteine Endopeptidases/chemistry , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Neoplasm Metastasis , Osteoclasts/cytology , T-Lymphocytes/cytology
12.
Chem Biol ; 22(8): 995-1001, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26256478

ABSTRACT

Bleomycin hydrolase (BLMH) is a neutral cysteine aminopeptidase that has been ascribed roles in many physiological and pathological processes, yet its primary biological function remains enigmatic. In this work, we describe the results of screening of a library of fluorogenic substrates to identify non-natural amino acids that are optimally recognized by BLMH. This screen identified several substrates with kcat/KM values that are substantially improved over the previously reported fluorogenic substrates for this enzyme. The substrate sequences were used to design activity-based probes that showed potent labeling of recombinant BLMH as well as endogenously expressed BLMH in cell extracts, and in intact cells. Importantly, we identify potent BLMH inhibitors that are able to fully inhibit endogenous BLMH activity in intact cells. These probes and inhibitors will be valuable new reagents to study BLMH function in cellular and animal models of human diseases where BLMH is likely to be involved.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Animals , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Drug Evaluation, Preclinical , Humans , Kinetics , Mice , Models, Molecular , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Structure-Activity Relationship , Substrate Specificity
13.
Haematologica ; 100(10): 1350-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26069288

ABSTRACT

Proteasome inhibitor resistance is a challenge for myeloma therapy. Bortezomib targets the ß5 and ß1 activity, but not the ß2 activity of the proteasome. Bortezomib-resistant myeloma cells down-regulate the activation status of the unfolded protein response, and up-regulate ß2 proteasome activity. To improve proteasome inhibition in bortezomib-resistant myeloma and to achieve more efficient UPR activation, we have developed LU-102, a selective inhibitor of the ß2 proteasome activity. LU-102 inhibited the ß2 activity in intact myeloma cells at low micromolar concentrations without relevant co-inhibition of ß1 and ß5 proteasome subunits. In proteasome inhibitor-resistant myeloma cells, significantly more potent proteasome inhibition was achieved by bortezomib or carfilzomib in combination with LU-102, compared to bortezomib/carfilzomib alone, resulting in highly synergistic cytotoxic activity of the drug combination via endoplasmatic reticulum stress-induced apoptosis. Combining bortezomib/carfilzomib with LU-102 significantly prolonged proteasome inhibition and increased activation of the unfolded protein response and IRE1-a activity. IRE1-α has recently been shown to control myeloma cell differentiation and bortezomib sensitivity (Leung-Hagesteijn, Cancer Cell 24:3, 289-304). Thus, ß2-selective proteasome inhibition by LU-102 in combination with bortezomib or carfilzomib results in synergistic proteasome inhibition, activation of the unfolded protein response, and cytotoxicity, and overcomes bortezomib/carfilzomib resistance in myeloma cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
14.
Mol Cell Proteomics ; 14(8): 2177-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041847

ABSTRACT

Tripeptidyl peptidase II (TPP2) is a serine peptidase involved in various biological processes, including antigen processing, cell growth, DNA repair, and neuropeptide mediated signaling. The underlying mechanisms of how a peptidase can influence this multitude of processes still remain unknown. We identified rapid proteomic changes in neuroblastoma cells following selective TPP2 inhibition using the known reversible inhibitor butabindide, as well as a new, more potent, and irreversible peptide phosphonate inhibitor. Our data show that TPP2 inhibition indirectly but rapidly decreases the levels of active, di-phosphorylated extracellular signal-regulated kinase 1 (ERK1) and ERK2 in the nucleus, thereby down-regulating signal transduction downstream of growth factors and mitogenic stimuli. We conclude that TPP2 mediates many important cellular functions by controlling ERK1 and ERK2 phosphorylation. For instance, we show that TPP2 inhibition of neurons in the hippocampus leads to an excessive strengthening of synapses, indicating that TPP2 activity is crucial for normal brain function.


Subject(s)
Aminopeptidases/metabolism , Cell Nucleus/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Serine Endopeptidases/metabolism , Aminopeptidases/antagonists & inhibitors , Animals , Cell Line , Cell Nucleus/drug effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Gene Ontology , Humans , Inhibitory Concentration 50 , Isotope Labeling , Mice , Models, Biological , Neurites/drug effects , Neurites/metabolism , Neuronal Plasticity/drug effects , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
15.
J Am Chem Soc ; 137(14): 4771-7, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25785540

ABSTRACT

The cysteine cathepsins are a group of 11 proteases whose function was originally believed to be the degradation of endocytosed material with a high degree of redundancy. However, it has become clear that these enzymes are also important regulators of both health and disease. Thus, selective tools that can discriminate between members of this highly related class of enzymes will be critical to further delineate the unique biological functions of individual cathepsins. Here we present the design and synthesis of a near-infrared quenched activity-based probe (qABP) that selectively targets cathepsin S which is highly expressed in immune cells. Importantly, this high degree of selectivity is retained both in vitro and in vivo. In combination with a new green-fluorescent pan-reactive cysteine cathepsin qABP we performed dual color labeling studies in bone marrow-derived immune cells and identified vesicles containing exclusively cathepsin S activity. This observation demonstrates the value of our complementary cathepsin probes and provides evidence for the existence of specific localization of cathepsin S activity in dendritic cells.


Subject(s)
Cathepsins/chemistry , Cathepsins/metabolism , Drug Design , Fluorescent Dyes/chemistry , Infrared Rays , Optical Imaging/methods , Animals , Color , Dendritic Cells/enzymology , Humans , Mammary Neoplasms, Experimental/enzymology , Mice , RAW 264.7 Cells , Substrate Specificity
16.
Chem Biol ; 22(1): 148-58, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25579207

ABSTRACT

Early detection of colonic polyps can prevent up to 90% of colorectal cancer deaths. Conventional colonoscopy readily detects the majority of premalignant lesions, which exhibit raised morphology. However, lesions that are flat and depressed are often undetected using this method. Therefore, there is a need for molecular-based contrast agents to improve detection rates over conventional colonoscopy. We evaluated a quenched fluorescent activity-based probe (qABP; BMV109) that targets multiple cysteine cathepsins that are overexpressed in intestinal dysplasia in a genetic model of spontaneous intestinal polyp formation and in a chemically induced model of colorectal carcinoma. We found that the qABP selectively targets cysteine cathepsins, resulting in high sensitivity and specificity for intestinal tumors in mice and humans. Additionally, the qABP can be administered by either intravenous injection or by local delivery to the colon, making it a highly valuable tool for improved detection of colorectal lesions using fluorescence-guided colonoscopy.


Subject(s)
Cathepsins/chemistry , Fluorescent Dyes/chemistry , Intestinal Neoplasms/diagnosis , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Carbocyanines/chemistry , Catalytic Domain , Cathepsins/metabolism , Colonoscopy , Disease Models, Animal , Fluorescent Dyes/metabolism , Humans , Immunohistochemistry , Intestinal Neoplasms/chemistry , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Polyps/chemistry , Polyps/diagnosis , Polyps/pathology , Sensitivity and Specificity
17.
ACS Chem Biol ; 9(8): 1869-76, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24918547

ABSTRACT

The ubiquitin-proteasome system (UPS) is a potential pathway for therapeutic intervention for pathogens such as Plasmodium, the causative agent of malaria. However, due to the essential nature of this proteolytic pathway, proteasome inhibitors must avoid inhibition of the host enzyme complex to prevent toxic side effects. The Plasmodium proteasome is poorly characterized, making rational design of inhibitors that induce selective parasite killing difficult. In this study, we developed a chemical probe that labels all catalytic sites of the Plasmodium proteasome. Using this probe, we identified several subunit selective small molecule inhibitors of the parasite enzyme complex. Treatment with an inhibitor that is specific for the ß5 subunit during blood stage schizogony led to a dramatic decrease in parasite replication while short-term inhibition of the ß2 subunit did not affect viability. Interestingly, coinhibition of both the ß2 and ß5 catalytic subunits resulted in enhanced parasite killing at all stages of the blood stage life cycle and reduced parasite levels in vivo to barely detectable levels. Parasite killing was achieved with overall low host toxicity, something that has not been possible with existing proteasome inhibitors. Our results highlight differences in the subunit dependency of the parasite and human proteasome, thus providing a strategy for development of potent antimalarial drugs with overall low host toxicity.


Subject(s)
Plasmodium/enzymology , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Catalytic Domain , Molecular Probes , Plasmodium/growth & development , Proteasome Endopeptidase Complex/drug effects
18.
J Am Chem Soc ; 135(39): 14726-30, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-23971698

ABSTRACT

The cysteine cathepsins are a family of proteases that play important roles in both normal cellular physiology and many human diseases. In cancer, the activity of many of the cysteine cathepsins is upregulated and can be exploited for tumor imaging. Here we present the design and synthesis of a new class of quenched fluorescent activity-based probes (qABPs) containing a phenoxymethyl ketone (PMK) electrophile. These reagents show enhanced in vivo properties and broad reactivity resulting in dramatically improved labeling and tumor imaging properties compared to those of previously reported ABPs.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/enzymology , Breast/pathology , Cysteine Proteases/analysis , Fluorescent Dyes/chemistry , Animals , Breast/enzymology , Breast Neoplasms/pathology , Cell Line , Cells, Cultured , Cysteine/metabolism , Cysteine Proteases/metabolism , Female , Fluorescent Dyes/chemical synthesis , Humans , Ketones/chemical synthesis , Ketones/chemistry , Mice , Optical Imaging/methods
19.
Nat Protoc ; 8(6): 1155-68, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23702832

ABSTRACT

Activity-based protein profiling (ABPP) is a functional proteomics technique for directly monitoring the expression of active enzymes in cell extracts and living cells. The technique relies on irreversible inhibitors equipped with reactive groups (warheads) that covalently attach to the active site of enzymes and fluorescent or affinity tags for imaging and purification purposes, respectively. Here, a high-throughput and robust protocol for high-resolution quantitative activity-based proteasome profiling is described. We use both panreactive and subunit-specific fluorescent activity-based probes (ABPs) to quantify the proteasome activity in living cells, in the presence or absence of the potent proteasome inhibitor bortezomib. Active proteasome subunits from cell lysates are affinity-purified via a biotinylated ABP. Purification from live cells involves a two-step ABP approach using a reagent with a cell-permeable azide-warhead and postlysis installation of biotin. By means of liquid chromatography-mass spectrometry (LC-MS)-based proteomics, we can accurately identify the enriched proteins and the active site peptides of the enzymes, and relatively quantify all the proteasome activities in one experiment. The fluorescence ABPP protocols takes 2-3 d, and approximately 8-10 d are needed to complete the entire protocol.


Subject(s)
Chromatography, Liquid/methods , Proteasome Endopeptidase Complex/metabolism , Proteins/analysis , Tandem Mass Spectrometry/methods , Fluorescence , Molecular Probe Techniques , Molecular Structure
20.
J Med Chem ; 56(3): 1262-75, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23320547

ABSTRACT

Proteasomes degrade the majority of proteins in mammalian cells by a concerted action of three distinct pairs of active sites. The chymotrypsin-like sites are targets of antimyeloma agents bortezomib and carfilzomib. Inhibitors of the trypsin-like site sensitize multiple myeloma cells to these agents. Here we describe systematic effort to develop inhibitors with improved potency and cell permeability, yielding azido-Phe-Leu-Leu-4-aminomethyl-Phe-methyl vinyl sulfone (4a, LU-102), and a fluorescent activity-based probe for this site. X-ray structures of 4a and related inhibitors complexed with yeast proteasomes revealed the structural basis for specificity. Nontoxic to myeloma cells when used as a single agent, 4a sensitized them to bortezomib and carfilzomib. This sensitizing effect was much stronger than the synergistic effects of histone acetylase inhibitors or additive effects of doxorubicin and dexamethasone, raising the possibility that combinations of inhibitors of the trypsin-like site with bortezomib or carfilzomib would have stronger antineoplastic activity than combinations currently used clinically.


Subject(s)
Amino Acids/chemistry , Cell Membrane Permeability , Proteasome Inhibitors/chemistry , Trypsin/chemistry , Cell Line , Drug Design , Humans , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL
...