Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 136(5): 2086-93, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24472041

ABSTRACT

Thioamide quenchers can be paired with compact fluorophores to design "turn-on" fluorescent protease substrates. We have used this method to study a variety of serine-, cysteine-, carboxyl-, and metallo-proteases, including trypsin, chymotrypsin, pepsin, thermolysin, papain, and calpain. Since thioamides quench some fluorophores red-shifted from those naturally occurring in proteins, this technique can be used for real time monitoring of protease activity in crude preparations of virtually any protease. We demonstrate the value of this method in three model applications: (1) characterization of papain enzyme kinetics using rapid-mixing experiments, (2) selective monitoring of cleavage at a single site in a peptide with multiple proteolytic sites, and (3) analysis of the specificity of an inhibitor of calpain in cell lysates.


Subject(s)
Enzyme Assays , Fluorescent Dyes/chemistry , Peptide Hydrolases/metabolism , Peptides/chemistry , Thioamides/chemistry , Animals , Cells, Cultured , Fibroblasts/enzymology , Mice , Spectrometry, Fluorescence , Substrate Specificity
2.
Cell Host Microbe ; 13(1): 15-28, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23332153

ABSTRACT

Following intracellular replication, the apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii cause host cell cytolysis to facilitate parasite release and disease progression. Parasite exit from infected cells requires the interplay of parasite-derived proteins and host actin cytoskeletal changes; however, the host proteins underlying these changes remain obscure. We report the identification of a Gα(q)-coupled host-signaling cascade required for the egress of both P. falciparum and T. gondii. Gα(q)-coupled signaling results in protein kinase C (PKC)-mediated loss of the host cytoskeletal protein adducin and weakening of the cellular cytoskeleton. This cytoskeletal compromise induces catastrophic Ca(2+) influx mediated by the mechanosensitive cation channel TRPC6, which activates host calpain that proteolyzes the host cytoskeleton allowing parasite release. Reinforcing the feasibility of targeting host proteins as an antiparasitic strategy, mammalian PKC inhibitors demonstrated activity in murine models of malaria and toxoplasmosis. Importantly, an orally bioavailable PKC inhibitor prolonged survival in an experimental cerebral malaria model.


Subject(s)
Autophagy , Host-Parasite Interactions , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Toxoplasma/physiology , Toxoplasmosis/metabolism , Animals , Autolysis , Calcium/metabolism , Cell Line , Cytoskeleton/metabolism , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/physiopathology , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Toxoplasmosis/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/physiopathology
3.
Proc Natl Acad Sci U S A ; 109(52): 21486-91, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236186

ABSTRACT

Early secretory and endoplasmic reticulum (ER)-localized proteins that are terminally misfolded or misassembled are degraded by a ubiquitin- and proteasome-mediated process known as ER-associated degradation (ERAD). Protozoan pathogens, including the causative agents of malaria, toxoplasmosis, trypanosomiasis, and leishmaniasis, contain a minimal ERAD network relative to higher eukaryotic cells, and, because of this, we observe that the malaria parasite Plasmodium falciparum is highly sensitive to the inhibition of components of this protein quality control system. Inhibitors that specifically target a putative protease component of ERAD, signal peptide peptidase (SPP), have high selectivity and potency for P. falciparum. By using a variety of methodologies, we validate that SPP inhibitors target P. falciparum SPP in parasites, disrupt the protein's ability to facilitate degradation of unstable proteins, and inhibit its proteolytic activity. These compounds also show low nanomolar activity against liver-stage malaria parasites and are also equipotent against a panel of pathogenic protozoan parasites. Collectively, these data suggest ER quality control as a vulnerability of protozoan parasites, and that SPP inhibition may represent a suitable transmission blocking antimalarial strategy and potential pan-protozoan drug target.


Subject(s)
Antiparasitic Agents/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Design , Endoplasmic Reticulum-Associated Degradation/drug effects , Protease Inhibitors/pharmacology , Animals , Antiparasitic Agents/chemistry , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Base Sequence , Computational Biology , Drug Resistance/drug effects , Endoplasmic Reticulum Stress/drug effects , Hep G2 Cells , Humans , Life Cycle Stages/drug effects , Liver/drug effects , Liver/parasitology , Molecular Sequence Data , Parasites/drug effects , Parasites/enzymology , Parasites/growth & development , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Protease Inhibitors/chemistry , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Proteome/metabolism , Small Molecule Libraries/pharmacology , Toxoplasma/drug effects , Toxoplasma/enzymology , Toxoplasma/growth & development , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/growth & development
4.
Cell Host Microbe ; 12(6): 815-23, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23245326

ABSTRACT

Plasmodium falciparum pathogenesis is affected by various cell types in the blood, including platelets, which can kill intraerythrocytic malaria parasites. Platelets could mediate these antimalarial effects through human defense peptides (HDPs), which exert antimicrobial effects by permeabilizing membranes. Therefore, we screened a panel of HDPs and determined that human platelet factor 4 (hPF4) kills malaria parasites inside erythrocytes by selectively lysing the parasite digestive vacuole (DV). PF4 rapidly accumulates only within infected erythrocytes and is required for parasite killing in infected erythrocyte-platelet cocultures. To exploit this antimalarial mechanism, we tested a library of small, nonpeptidic mimics of HDPs (smHDPs) and identified compounds that kill P. falciparum by rapidly lysing the parasite DV while sparing the erythrocyte plasma membrane. Lead smHDPs also reduced parasitemia in a murine malaria model. Thus, identifying host molecules that control parasite growth can further the development of related molecules with therapeutic potential.


Subject(s)
Antimalarials/isolation & purification , Antimalarials/metabolism , Plasmodium falciparum/drug effects , Platelet Factor 4/metabolism , Animals , Cell Survival/drug effects , Disease Models, Animal , Erythrocytes/parasitology , Malaria/drug therapy , Malaria/parasitology , Mice , Parasite Load , Parasitemia/drug therapy , Parasitemia/parasitology
5.
J Am Chem Soc ; 134(42): 17704-13, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-22998171

ABSTRACT

We have designed a highly specific inhibitor of calpain by mimicking a natural protein-protein interaction between calpain and its endogenous inhibitor calpastatin. To enable this goal we established a new method of stabilizing an α-helix in a small peptide by screening 24 commercially available cross-linkers for successful cysteine alkylation in a model peptide sequence. The effects of cross-linking on the α-helicity of selected peptides were examined by CD and NMR spectroscopy, and revealed structurally rigid cross-linkers to be the best at stabilizing α-helices. We applied this strategy to the design of inhibitors of calpain that are based on calpastatin, an intrinsically unstable polypeptide that becomes structured upon binding to the enzyme. A two-turn α-helix that binds proximal to the active site cleft was stabilized, resulting in a potent and selective inhibitor for calpain. We further expanded the utility of this inhibitor by developing irreversible calpain family activity-based probes (ABPs), which retained the specificity of the stabilized helical inhibitor. We believe the inhibitor and ABPs will be useful for future investigation of calpains, while the cross-linking technique will enable exploration of other protein-protein interactions.


Subject(s)
Calcium-Binding Proteins/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Calcium-Binding Proteins/chemical synthesis , Calcium-Binding Proteins/chemistry , Calpain/chemistry , Calpain/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Secondary , Structure-Activity Relationship
6.
J Biol Chem ; 287(20): 16860-8, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22433849

ABSTRACT

Host-derived proteases are crucial for the successful infection of vertebrates by several pathogens, including the Lyme disease spirochete bacterium, Borrelia burgdorferi. B. burgdorferi must traverse tissue barriers in the tick vector during transmission to the host and during dissemination within the host, and it must disrupt immune challenges to successfully complete its infectious cycle. It has been proposed that B. burgdorferi can accomplish these tasks without an endogenous extra-cytoplasmic protease by commandeering plasminogen, the highly abundant precursor of the vertebrate protease plasmin. However, the molecular mechanism by which B. burgdorferi immobilizes plasminogen to its surface remains obscure. The data presented here demonstrate that the outer surface protein C (OspC) of B. burgdorferi is a potent plasminogen receptor on the outer membrane of the bacterium. OspC-expressing spirochetes readily bind plasminogen, whereas only background levels of plasminogen are detectable on OspC-deficient strains. Furthermore, plasminogen binding by OspC-expressing spirochetes can be significantly reduced using anti-OspC antibodies. Co-immunofluorescence staining assays demonstrate that wild-type bacteria immobilize plasminogen only if they are actively expressing OspC regardless of the expression of other surface proteins. The co-localization of plasminogen and OspC on OspC-expressing spirochetes further implicates OspC as a biologically relevant plasminogen receptor on the surface of live B. burgdorferi.


Subject(s)
Antigens, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/biosynthesis , Borrelia burgdorferi/metabolism , Gene Expression Regulation, Bacterial/physiology , Lyme Disease/metabolism , Plasminogen/metabolism , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/agonists , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Humans , Lyme Disease/genetics , Plasminogen/genetics
7.
Biochimie ; 94(2): 318-27, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21802486

ABSTRACT

Human strains of Staphylococcus aureus secrete two papain-like proteases, staphopain A and B. Avian strains produce another homologous enzyme, staphopain C. Animal studies suggest that staphopains B and C contribute to bacterial virulence, in contrast to staphopain A, which seems to have a virulence unrelated function. Here we present a detailed study of substrate preferences of all three proteases. The specificity of staphopain A, B and C substrate-binding subsites was mapped using different synthetic substrate libraries, inhibitor libraries and a protein substrate combinatorial library. The analysis demonstrated that the most efficiently hydrolyzed sites, using Schechter and Berger nomenclature, comprise a P2-Gly↓Ala(Ser) sequence motif, where P2 distinguishes the specificity of staphopain A (Leu) from that of both staphopains B and C (Phe/Tyr). However, we show that at the same time the overall specificity of staphopains is relaxed, insofar as multiple substrates that diverge from the sequences described above are also efficiently hydrolyzed.


Subject(s)
Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Proteases/metabolism , Staphylococcus aureus , Amino Acid Motifs , Animals , Bacterial Proteins/isolation & purification , Binding, Competitive , Birds , Catalytic Domain , Cysteine Endopeptidases/isolation & purification , Humans , Hydrolysis , Kinetics , Molecular Sequence Data , Protein Binding , Small Molecule Libraries , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Substrate Specificity , Virulence
8.
PLoS Pathog ; 7(9): e1002139, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21909255

ABSTRACT

Trypanosoma cruzi is the causative agent of Chagas' disease. Novel chemotherapy with the drug K11777 targets the major cysteine protease cruzain and disrupts amastigote intracellular development. Nevertheless, the biological role of the protease in infection and pathogenesis remains unclear as cruzain gene knockout failed due to genetic redundancy. A role for the T. cruzi cysteine protease cruzain in immune evasion was elucidated in a comparative study of parental wild type- and cruzain-deficient parasites. Wild type T. cruzi did not activate host macrophages during early infection (<60 min) and no increase in ∼P iκB was detected. The signaling factor NF-κB P65 colocalized with cruzain on the cell surface of intracellular wild type parasites, and was proteolytically cleaved. No significant IL-12 expression occurred in macrophages infected with wild type T. cruzi and treated with LPS and BFA, confirming impairment of macrophage activation pathways. In contrast, cruzain-deficient parasites induced macrophage activation, detectable iκB phosphorylation, and nuclear NF-κB P65 localization. These parasites were unable to develop intracellularly and survive within macrophages. IL 12 expression levels in macrophages infected with cruzain-deficient T. cruzi were comparable to LPS activated controls. Thus cruzain hinders macrophage activation during the early (<60 min) stages of infection, by interruption of the NF-κB P65 mediated signaling pathway. These early events allow T. cruzi survival and replication, and may lead to the spread of infection in acute Chagas' disease.


Subject(s)
Cysteine Endopeptidases/physiology , Immune Evasion/physiology , Macrophages/parasitology , Protozoan Proteins/physiology , Animals , Arginase/biosynthesis , Cysteine Endopeptidases/deficiency , Dipeptides/pharmacology , Humans , I-kappa B Proteins/metabolism , Interleukin-12/biosynthesis , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Phenylalanine/analogs & derivatives , Piperazines , Tosyl Compounds , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/genetics , Vinyl Compounds/pharmacology
9.
Mol Cell Proteomics ; 10(12): M111.010678, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21903871

ABSTRACT

Plasmodium falciparum is an obligate intracellular pathogen responsible for worldwide morbidity and mortality. This parasite establishes a parasitophorous vacuole within infected red blood cells wherein it differentiates into multiple daughter cells that must rupture their host cells to continue another infectious cycle. Using atomic force microscopy, we establish that progressive macrostructural changes occur to the host cell cytoskeleton during the last 15 h of the erythrocytic life cycle. We used a comparative proteomics approach to determine changes in the membrane proteome of infected red blood cells during the final steps of parasite development that lead to egress. Mass spectrometry-based analysis comparing the red blood cell membrane proteome in uninfected red blood cells to that of infected red blood cells and postrupture vesicles highlighted two temporally distinct events; (Hay, S. I., et al. (2009). A world malaria map: Plasmodium falciparum endemicity in 2007. PLoS Med. 6, e1000048) the striking loss of cytoskeletal adaptor proteins that are part of the junctional complex, including α/ß-adducin and tropomyosin, correlating temporally with the emergence of large holes in the cytoskeleton seen by AFM as early ~35 h postinvasion, and (Maier, A. G., et al. (2008) Exported proteins required for virulence and rigidity of Plasmodium falciparum-infected human erythrocytes. Cell 134, 48-61) large-scale proteolysis of the cytoskeleton during rupture ~48 h postinvasion, mediated by host calpain-1. We thus propose a sequential mechanism whereby parasites first remove a selected set of cytoskeletal adaptor proteins to weaken the host membrane and then use host calpain-1 to dismantle the remaining cytoskeleton, leading to red blood cell membrane collapse and parasite release.


Subject(s)
Cytoskeleton/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Plasmodium falciparum/physiology , Calmodulin-Binding Proteins/metabolism , Calpain/metabolism , Cell Membrane/metabolism , Cell Membrane/parasitology , Cells, Cultured , Cytoskeletal Proteins/metabolism , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Merozoites , Microscopy, Atomic Force , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proteolysis , Tandem Mass Spectrometry
10.
Proc Natl Acad Sci U S A ; 108(34): E526-34, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21844374

ABSTRACT

Malaria causes worldwide morbidity and mortality, and while chemotherapy remains an excellent means of malaria control, drug-resistant parasites necessitate the discovery of new antimalarials. Peptidases are a promising class of drug targets and perform several important roles during the Plasmodium falciparum erythrocytic life cycle. Herein, we report a multidisciplinary effort combining activity-based protein profiling, biochemical, and peptidomic approaches to functionally analyze two genetically essential P. falciparum metallo-aminopeptidases (MAPs), PfA-M1 and Pf-LAP. Through the synthesis of a suite of activity-based probes (ABPs) based on the general MAP inhibitor scaffold, bestatin, we generated specific ABPs for these two enzymes. Specific inhibition of PfA-M1 caused swelling of the parasite digestive vacuole and prevented proteolysis of hemoglobin (Hb)-derived oligopeptides, likely starving the parasite resulting in death. In contrast, inhibition of Pf-LAP was lethal to parasites early in the life cycle, prior to the onset of Hb degradation suggesting that Pf-LAP has an essential role outside of Hb digestion.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Leucine/analogs & derivatives , Malaria/parasitology , Molecular Probe Techniques , Molecular Probes/metabolism , Multigene Family , Amino Acid Sequence , Aminopeptidases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hemoglobins/metabolism , Leucine/chemistry , Leucine/pharmacology , Leucyl Aminopeptidase/antagonists & inhibitors , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides/chemistry , Peptides/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Array Analysis , Protein Processing, Post-Translational/drug effects , Substrate Specificity/drug effects
11.
J Med Chem ; 54(6): 1655-66, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21366301

ABSTRACT

The malarial PfA-M1 metallo-aminopeptidase is considered a putative drug target. The natural product dipeptide mimetic, bestatin, is a potent inhibitor of PfA-M1. Herein we present a new, efficient, and high-yielding protocol for the synthesis of bestatin derivatives from natural and unnatural N-Boc-d-amino acids. A diverse library of bestatin derivatives was synthesized with variants at the side chain of either the α-hydroxy-ß-amino acid (P1) or the adjacent natural α-amino acid (P1'). Surprisingly, we found that extended aromatic side chains at the P1 position resulted in potent inhibition against PfA-M1. To understand these data, we determined the X-ray cocrystal structures of PfA-M1 with two derivatives having either a Tyr(OMe) 15 or Tyr(OBzl) 16 at the P1 position and observed substantial inhibitor-induced rearrangement of the primary loop within the PfA-M1 pocket that interacts with the P1 side chain. Our data provide important insights for the rational design of more potent and selective inhibitors of this enzyme that may eventually lead to new therapies for malaria.


Subject(s)
Antimalarials/chemical synthesis , CD13 Antigens/antagonists & inhibitors , Leucine/analogs & derivatives , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Antimalarials/pharmacology , Binding Sites , CD13 Antigens/chemistry , Crystallography, X-Ray , Leucine/chemical synthesis , Leucine/chemistry , Leucine/pharmacology , Models, Molecular , Molecular Structure , Plasmodium falciparum/enzymology , Protein Binding , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship
12.
Antimicrob Agents Chemother ; 54(9): 3597-604, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20547797

ABSTRACT

Malaria is a global health problem that causes significant mortality and morbidity, with more than 1 million deaths per year caused by Plasmodium falciparum. Most antimalarial drugs face decreased efficacy due to the emergence of resistant parasites, which necessitates the discovery of new drugs. To identify new antimalarials, we developed an automated 384-well plate screening assay using P. falciparum parasites that stably express cytoplasmic firefly luciferase. After initial optimization, we tested two different types of compound libraries: known bioactive collections (Library of Pharmacologically Active Compounds [LOPAC] and the library from the National Institute of Neurological Disorders and Stroke [NINDS]) and a library of uncharacterized compounds (ChemBridge). A total of 12,320 compounds were screened at 5.5 microM. Selecting only compounds that reduced parasite growth by 85% resulted in 33 hits from the combined bioactive collection and 130 hits from the ChemBridge library. Fifteen novel drug-like compounds from the bioactive collection were found to be active against P. falciparum. Twelve new chemical scaffolds were found from the ChemBridge hits, the most potent of which was a series based on the 1,4-naphthoquinone scaffold, which is structurally similar to the FDA-approved antimalarial atovaquone. However, in contrast to atovaquone, which acts to inhibit the bc(1) complex and block the electron transport chain in parasite mitochondria, we have determined that our new 1,4-napthoquinones act in a novel, non-bc(1)-dependent mechanism and remain potent against atovaquone- and chloroquine-resistant parasites. Ultimately, this study may provide new probes to understand the molecular details of the malaria life cycle and to identify new antimalarials.


Subject(s)
Antimalarials/pharmacology , Drug Resistance, Multiple/drug effects , Parasitic Sensitivity Tests/methods , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemistry , Molecular Structure
13.
Mol Pharmacol ; 78(2): 319-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466822

ABSTRACT

A tetrahydroquinoline oxocarbazate (PubChem CID 23631927) was tested as an inhibitor of human cathepsin L (EC 3.4.22.15) and as an entry blocker of severe acute respiratory syndrome (SARS) coronavirus and Ebola pseudotype virus. In the cathepsin L inhibition assay, the oxocarbazate caused a time-dependent 17-fold drop in IC(50) from 6.9 nM (no preincubation) to 0.4 nM (4-h preincubation). Slowly reversible inhibition was demonstrated in a dilution assay. A transient kinetic analysis using a single-step competitive inhibition model provided rate constants of k(on) = 153,000 M(-1)s(-1) and k(off) = 4.40 x 10(-5) s(-1) (K(i) = 0.29 nM). The compound also displayed cathepsin L/B selectivity of >700-fold and was nontoxic to human aortic endothelial cells at 100 muM. The oxocarbazate and a related thiocarbazate (PubChem CID 16725315) were tested in a SARS coronavirus (CoV) and Ebola virus-pseudotype infection assay with the oxocarbazate but not the thiocarbazate, demonstrating activity in blocking both SARS-CoV (IC(50) = 273 +/- 49 nM) and Ebola virus (IC(50) = 193 +/- 39 nM) entry into human embryonic kidney 293T cells. To trace the intracellular action of the inhibitors with intracellular cathepsin L, the activity-based probe biotin-Lys-C5 alkyl linker-Tyr-Leu-epoxide (DCG-04) was used to label the active site of cysteine proteases in 293T lysates. The reduction in active cathepsin L in inhibitor-treated cells correlated well with the observed potency of inhibitors observed in the virus pseudotype infection assay. Overall, the oxocarbazate CID 23631927 was a subnanomolar, slow-binding, reversible inhibitor of human cathepsin L that blocked SARS-CoV and Ebola pseudotype virus entry in human cells.


Subject(s)
Antiviral Agents/pharmacology , Cathepsin L/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Hemorrhagic Fever, Ebola/prevention & control , Quinolines/pharmacology , Quinolones/pharmacology , Severe Acute Respiratory Syndrome/prevention & control , Cell Line , Humans , Kinetics
15.
Semin Cell Dev Biol ; 20(2): 225-30, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19429495

ABSTRACT

Five genes encode the five human signal peptide peptidases (SPPs), which are intramembrane-cleaving aspartyl proteases (aspartyl I-CLiPs). SPPs have been conserved through evolution with family members found in higher eukaryotes, fungi, protozoa, arachea, and plants. SPPs are related to the presenilin family of aspartyl I-CLiPs but differ in several key aspects. Presenilins (PSENs) and SPPs both cleave the transmembrane region of membrane proteins; however, PSENs cleave type 1 membrane proteins whereas SPPs cleave type 2 membrane proteins. Though the overall homology between SPPs and PSENs is minimal, they are multipass membrane proteins that contain two conserved active site motifs YD and GxGD in adjacent membrane-spanning domains and a conserved PAL motif of unknown function near their COOH-termini. They differ in that the active site YD and GxGD containing transmembrane domains of SPPs are inverted relative to PSENs, thus, orienting the active site in a consistent topology relative to the substrate. At least two of the human SPPs (SPP and SPPL3) appear to function without additional cofactors, but PSENs function as a protease, called gamma-secretase, only when complexed with Nicastrin, APH-1 and Pen-2. The biological roles of SPP are largely unknown, and only a few endogenous substrates for SPPs have been identified. Nevertheless there is emerging evidence that SPP family members are highly druggable and may regulate both essential physiologic and pathophysiologic processes. Further study of the SPP family is needed in order to understand their biological roles and their potential as therapeutic targets.


Subject(s)
Aspartic Acid Endopeptidases/classification , Receptor, PAR-2/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/pharmacology , Humans , Models, Biological , Phylogeny
16.
Science ; 324(5928): 794-7, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19342550

ABSTRACT

Apicomplexan parasites, including Plasmodium falciparum and Toxoplasma gondii (the causative agents of malaria and toxoplasmosis, respectively), are responsible for considerable morbidity and mortality worldwide. These pathogenic protozoa replicate within an intracellular vacuole inside of infected host cells, from which they must escape to initiate a new lytic cycle. By integrating cell biological, pharmacological, and genetic approaches, we provide evidence that both Plasmodium and Toxoplasma hijack host cell calpain proteases to facilitate parasite egress. Immunodepletion or inhibition of calpain-1 in hypotonically lysed and resealed erythrocytes prevented the escape of P. falciparum parasites, which was restored by adding purified calpain-1. Similarly, efficient egress of T. gondii from mammalian fibroblasts was blocked by either small interfering RNA-mediated suppression or genetic deletion of calpain activity and could be restored by genetic complementation.


Subject(s)
Calpain/metabolism , Erythrocytes/parasitology , Plasmodium falciparum/pathogenicity , Toxoplasma/pathogenicity , Animals , Calpain/blood , Calpain/genetics , Cell Line , Cell Line, Tumor , Fibroblasts/parasitology , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Life Cycle Stages , Merozoites/physiology , Mice , Mice, Knockout , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , RNA, Small Interfering , Schizonts/physiology , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasma/physiology
17.
Bioorg Med Chem Lett ; 18(22): 5932-6, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18823778

ABSTRACT

A novel set of activity-based probes (ABPs) for functionally profiling metallo-aminopeptidases was synthesized based on the bestatin inhibitor scaffold, the first synthesis of bestatin analogues using solid-phase techniques. These ABPs were shown to label metallo-aminopeptidases, using both a biotin and a fluorophore reporter, in an activity-dependent manner. This probe class was also shown to be amenable to 'click' chemistry labeling for possible use in live cells. Finally, we demonstrate that the ABPs are able to label an aminopeptidase in a complex proteome. Thus, these bestatin-based probes should have wide utility to functionally profile aminopeptidases in many biological systems.


Subject(s)
Aminopeptidases/metabolism , Leucine/analogs & derivatives , Models, Molecular , Catalytic Domain , Fluorescent Dyes , Leucine/pharmacology , Molecular Structure , Structure-Activity Relationship
18.
Trends Pharmacol Sci ; 29(2): 51-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199489

ABSTRACT

Malaria is a global disease, causing at least 500 million clinical cases and more than one million deaths each year. Moreover, drug-resistant Plasmodium falciparum, the organism that causes most malaria-associated deaths, has become a major problem. Therefore, discovery and investigation of novel targets for anti-malarial drug design is essential to combat this disease. The malarial genome has been sequenced, revealing approximately 5500 genes. The current post-genomic challenge is functionally to evaluate the essential genes and validate them for therapeutic design. Unfortunately, standard genetics techniques are limited in scope because of low transfection efficiency and a lack of knockdown techniques, thereby rendering the analysis of essential genes difficult.


Subject(s)
Drug Delivery Systems , Genetic Techniques , Malaria, Falciparum/drug therapy , Animals , Antimalarials/pharmacology , Drug Design , Drug Resistance , Humans , Malaria, Falciparum/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics
19.
Mol Biochem Parasitol ; 154(2): 148-57, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553576

ABSTRACT

Merozoite surface proteins of the human malaria parasite Plasmodium falciparum are involved in initial contact with target erythrocytes, a process that begins a cascade of events required for successful invasion of these cells. In order to identify complexes that may play a role in invasion we purified detergent-resistant membranes (DRMs), known to be enriched in merozoite surface proteins, and used blue native-polyacrylamide gel electrophoresis (BN-PAGE) to isolate high molecular weight complexes for identification by mass spectrometry. Sixty-two proteins were detected and these mostly belonged to expected DRM proteins classes including GPI-anchored, multi-membrane spanning and rhoptry proteins. Proteins from seven known complexes were identified including MSP-1/7, the low (RAP1/2 and RAP1/3), and high (RhopH1/H2/H3) molecular weight rhoptry complexes, and the invasion motor complex (GAP45/GAP50/myosinA). Remarkably, a large proportion of identified spectra were derived from only 4 proteins: the GPI-anchored proteins MSP-1 and Pf92, the putative GPI-anchored protein Pf113 and RAP-1, the core component of the two RAP complexes. Each of these proteins predominated in high molecular weight species suggesting their aggregation in much larger complexes than anticipated. To demonstrate that the procedure had isolated novel complexes we focussed on MSP-1, which predominated as a distinct species at approximately 500 kDa by BN-PAGE, approximately twice its expected size. Chemical cross-linking supports the existence of a stable MSP-1 oligomer of approximately 500 kDa, probably comprising a highly stable homodimeric species. Our observations also suggests that oligomerization of MSP-1 is likely to occur outside the C-terminal epidermal growth factor (EGF)-like domains. Confirmation of MSP-1 oligomerization, together with the isolation of a number of known complexes by BN-PAGE, makes it highly likely that novel interactions occur amongst members of this proteome.


Subject(s)
Detergents/pharmacology , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Proteome/isolation & purification , Protozoan Proteins/isolation & purification , Animals , Electrophoresis, Gel, Two-Dimensional , Merozoite Surface Protein 1/chemistry , Merozoite Surface Protein 1/isolation & purification , Molecular Weight , Schizonts/chemistry , Schizonts/drug effects
20.
J Biol Chem ; 281(18): 12824-32, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16520377

ABSTRACT

The substrate specificities of papain-like cysteine proteases (clan CA, family C1) papain, bromelain, and human cathepsins L, V, K, S, F, B, and five proteases of parasitic origin were studied using a completely diversified positional scanning synthetic combinatorial library. A bifunctional coumarin fluorophore was used that facilitated synthesis of the library and individual peptide substrates. The library has a total of 160,000 tetrapeptide substrate sequences completely randomizing each of the P1, P2, P3, and P4 positions with 20 amino acids. A microtiter plate assay format permitted a rapid determination of the specificity profile of each enzyme. Individual peptide substrates were then synthesized and tested for a quantitative determination of the specificity of the human cathepsins. Despite the conserved three-dimensional structure and similar substrate specificity of the enzymes studied, distinct amino acid preferences that differentiate each enzyme were identified. The specificities of cathepsins K and S partially match the cleavage site sequences in their physiological substrates. Capitalizing on its unique preference for proline and glycine at the P2 and P3 positions, respectively, selective substrates and a substrate-based inhibitor were developed for cathepsin K. A cluster analysis of the proteases based on the complete specificity profile provided a functional characterization distinct from standard sequence analysis. This approach provides useful information for developing selective chemical probes to study protease-related pathologies and physiologies.


Subject(s)
Peptides/chemistry , Animals , Binding, Competitive , Cathepsin K , Cathepsins/chemistry , Coumarins/chemistry , Cysteine/chemistry , Humans , Ketones/chemistry , Kinetics , Models, Chemical , Phylogeny , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...