Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Publication year range
1.
J Proteome Res ; 18(5): 2078-2087, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30964683

ABSTRACT

Purines represent a class of essential metabolites produced by the cell to maintain cellular homeostasis and facilitate cell proliferation. In times of high purine demand, the de novo purine biosynthetic pathway is activated; however, the mechanisms that facilitate this process are largely unknown. One plausible mechanism is through intracellular signaling, which results in enzymes within the pathway becoming post-translationally modified to enhance their individual enzyme activities and the overall pathway metabolic flux. Here, we employ a proteomic strategy to investigate the extent to which de novo purine biosynthetic pathway enzymes are post-translationally modified in 293T cells. We identified 7 post-translational modifications on 135 residues across the 6 human pathway enzymes. We further asked whether there were differences in the post-translational modification state of each pathway enzyme isolated from cells cultured in the presence or absence of purines. Of the 174 assigned modifications, 67% of them were only detected in one experimental growth condition in which a significant number of serine and threonine phosphorylations were noted. A survey of the most-probable kinases responsible for these phosphorylation events uncovered a likely AKT phosphorylation site at residue Thr397 of PPAT, which was only detected in cells under purine-supplemented growth conditions. These data suggest that this modification might alter enzyme activity or modulate its interaction(s) with downstream pathway enzymes. Together, these findings propose a role for post-translational modifications in pathway regulation and activation to meet intracellular purine demand.


Subject(s)
Amidophosphoribosyltransferase/metabolism , Peptide Mapping/methods , Protein Processing, Post-Translational , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Purines/metabolism , Acetylation , Adenylosuccinate Lyase/genetics , Adenylosuccinate Lyase/metabolism , Amidophosphoribosyltransferase/genetics , Amino Acid Sequence , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Peptides/chemical synthesis , Peptides/metabolism , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Signal Transduction , Threonine/metabolism , Ubiquitination
2.
Biol Chem ; 400(12): 1629-1638, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31256057

ABSTRACT

Incidental detection of pancreatic cysts has increased dramatically over the last decade, but risk stratification and clinical management remain a challenge. Mucinous cysts are precursor lesions to pancreatic cancer, however, the majority are indolent. Current diagnostics cannot identify mucinous cysts that harbor cancer or reliably differentiate these lesions from nonmucinous cysts, which present minimal risk of malignant progression. We previously determined that activity of two aspartyl proteases was increased in mucinous cysts. Using a global protease activity profiling technology, termed multiplex substrate profiling by mass spectrometry (MSP-MS), we now show that aminopeptidase activity is also elevated in mucinous cysts. The serine aminopeptidase, tripeptidyl peptidase 1 (TPP1), was detected by proteomic analysis of cyst fluid samples and quantitation using targeted MS demonstrated that this protease was significantly more abundant in mucinous cysts. In a cohort of 110 cyst fluid samples, TPP1 activity was increased more than 3-fold in mucinous cysts relative to nonmucinous cysts. Moreover, TPP1 activity is primarily associated with mucinous cysts that harbor high-grade dysplasia or invasive carcinoma. Although only 59% accurate for differentiating these lesions, measurement of TPP1 activity may improve early detection and treatment of high-risk pancreatic cysts when used in conjunction with other promising biomarkers.


Subject(s)
Aminopeptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Lysosomes/enzymology , Pancreatic Cyst/metabolism , Pancreatic Neoplasms/metabolism , Serine Proteases/metabolism , Humans , Lysosomes/metabolism , Pancreatic Cyst/pathology , Pancreatic Neoplasms/pathology , Proteomics , Tripeptidyl-Peptidase 1
3.
Proc Natl Acad Sci U S A ; 113(14): E2001-10, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27006500

ABSTRACT

Proteases constitute the largest enzyme family, yet their biological roles are obscured by our rudimentary understanding of their cellular substrates. There are 12 human caspases that play crucial roles in inflammation and cell differentiation and drive the terminal stages of cell death. Recent N-terminomics technologies have begun to enumerate the diverse substrates individual caspases can cleave in complex cell lysates. It is clear that many caspases have shared substrates; however, few data exist about the catalytic efficiencies (kcat/KM) of these substrates, which is critical to understanding their true substrate preferences. In this study, we use quantitative MS to determine the catalytic efficiencies for hundreds of natural protease substrates in cellular lysate for two understudied members: caspase-2 and caspase-6. Most substrates are new, and the cleavage rates vary up to 500-fold. We compare the cleavage rates for common substrates with those found for caspase-3, caspase-7, and caspase-8, involved in apoptosis. There is little correlation in catalytic efficiencies among the five caspases, suggesting each has a unique set of preferred substrates, and thus more specialized roles than previously understood. We synthesized peptide substrates on the basis of protein cleavage sites and found similar catalytic efficiencies between the protein and peptide substrates. These data suggest the rates of proteolysis are dominated more by local primary sequence, and less by the tertiary protein fold. Our studies highlight that global quantitative rate analysis for posttranslational modification enzymes in complex milieus for native substrates is critical to better define their functions and relative sequence of events.


Subject(s)
Caspases/metabolism , Mass Spectrometry/methods , Proteins/metabolism , Substrate Specificity
4.
BMC Genomics ; 19(1): 204, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29554870

ABSTRACT

BACKGROUND: Human herpesvirus-6A and -6B (HHV-6) are betaherpesviruses that reach > 90% seroprevalence in the adult population. Unique among human herpesviruses, HHV-6 can integrate into the subtelomeric regions of human chromosomes; when this occurs in germ line cells it causes a condition called inherited chromosomally integrated HHV-6 (iciHHV-6). Only two complete genomes are available for replicating HHV-6B, leading to numerous conflicting annotations and little known about the global genomic diversity of this ubiquitous virus. RESULTS: Using a custom capture panel for HHV-6B, we report complete genomes from 61 isolates of HHV-6B from active infections (20 from Japan, 35 from New York state, and 6 from Uganda), and 64 strains of iciHHV-6B (mostly from North America). HHV-6B sequence clustered by geography and illustrated extensive recombination. Multiple iciHHV-6B sequences from unrelated individuals across the United States were found to be completely identical, consistent with a founder effect. Several iciHHV-6B strains clustered with strains from recent active pediatric infection. Combining our genomic analysis with the first RNA-Seq and shotgun proteomics studies of HHV-6B, we completely reannotated the HHV-6B genome, altering annotations for more than 10% of existing genes, with multiple instances of novel splicing and genes that hitherto had gone unannotated. CONCLUSION: Our results are consistent with a model of intermittent de novo integration of HHV-6B into host germline cells during active infection with a large contribution of founder effect in iciHHV-6B. Our data provide a significant advance in the genomic annotation of HHV-6B, which will contribute to the detection, diversity, and control of this virus.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/metabolism , Roseolovirus Infections/virology , Viral Proteins/analysis , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Female , Genome, Viral , Global Health , Herpesvirus 6, Human/isolation & purification , Humans , Infant , Infant, Newborn , Male , Middle Aged , Proteomics/methods , Young Adult
5.
Biol Chem ; 399(12): 1375-1388, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30367778

ABSTRACT

Pseudogymnoascus destructans is a pathogenic fungus responsible for White-nose Syndrome (WNS), a disease afflicting multiple species of North American bats. Pseudogymnoascus destructans infects susceptible bats during hibernation, invading dermal tissue and causing extensive tissue damage. In contrast, other Pseudogymnoascus species are non-pathogenic and cross-species comparisons may therefore reveal factors that contribute to virulence. In this study, we compared the secretome of P. destructans with that from several closely related Pseudogymnoascus species. A diverse set of hydrolytic enzymes were identified, including a putative serine peptidase, PdCP1, that was unique to the P. destructans secretome. A recombinant form of PdCP1 was purified and substrate preference determined using a multiplexed-substrate profiling method based on enzymatic degradation of a synthetic peptide library and analysis by mass spectrometry. Most peptide substrates were sequentially truncated from the carboxyl-terminus revealing that this enzyme is a bona fide carboxypeptidase. Peptides with arginine located close to the carboxyl-terminus were rapidly cleaved, and a fluorescent substrate containing arginine was therefore used to characterize PdCP1 activity and to screen a selection of peptidase inhibitors. Antipain and leupeptin were found to be the most potent inhibitors of PdCP1 activity.


Subject(s)
Ascomycota/enzymology , Carboxypeptidases/metabolism , Chiroptera/microbiology , Mycoses/metabolism , Animals , Antipain/pharmacology , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/isolation & purification , Leupeptins/pharmacology , Mycoses/microbiology , Syndrome
6.
Proc Natl Acad Sci U S A ; 112(24): 7478-83, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-25944934

ABSTRACT

Pseudogymnoascus destructans is the causative agent of white-nose syndrome, a disease that has caused the deaths of millions of bats in North America. This psychrophilic fungus proliferates at low temperatures and targets hibernating bats, resulting in their premature arousal from stupor with catastrophic consequences. Despite the impact of white-nose syndrome, little is known about the fungus itself or how it infects its mammalian host. P. destructans is not amenable to genetic manipulation, and therefore understanding the proteins involved in infection requires alternative approaches. Here, we identify hydrolytic enzymes secreted by P. destructans, and use a novel and unbiased substrate profiling technique to define active peptidases. These experiments revealed that endopeptidases are the major proteolytic activities secreted by P. destructans, and that collagen, the major structural protein in mammals, is actively degraded by the secretome. A serine endopeptidase, hereby-named Destructin-1, was subsequently identified, and a recombinant form overexpressed and purified. Biochemical analysis of Destructin-1 showed that it mediated collagen degradation, and a potent inhibitor of peptidase activity was identified. Treatment of P. destructans-conditioned media with this antagonist blocked collagen degradation and facilitated the detection of additional secreted proteolytic activities, including aminopeptidases and carboxypeptidases. These results provide molecular insights into the secretome of P. destructans, and identify serine endopeptidases that have the clear potential to facilitate tissue invasion and pathogenesis in the mammalian host.


Subject(s)
Ascomycota/enzymology , Ascomycota/pathogenicity , Chiroptera/microbiology , Collagenases/metabolism , Fungal Proteins/metabolism , Mycoses/veterinary , Amino Acid Sequence , Animals , Ascomycota/genetics , Base Sequence , Catalytic Domain , Collagenases/chemistry , Collagenases/genetics , DNA, Fungal/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mycoses/microbiology , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Virulence
7.
Anal Chem ; 89(8): 4550-4558, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28322550

ABSTRACT

The more than 500 protein kinases comprising the human kinome catalyze hundreds of thousands of phosphorylation events to regulate a diversity of cellular functions; however, the extended substrate specificity is still unknown for many of these kinases. We report here a method for quantitatively describing kinase substrate specificity using an unbiased peptide library-based approach with direct measurement of phosphorylation by tandem liquid chromatography-tandem mass spectrometry (LC-MS/MS) peptide sequencing (multiplex substrate profiling by mass spectrometry, MSP-MS). This method can be deployed with as low as 10 nM enzyme to determine activity against S/T/Y-containing peptides; additionally, label-free quantitation is used to ascertain catalytic efficiency values for individual peptide substrates in the multiplex assay. Using this approach we developed quantitative motifs for a selection of kinases from each branch of the kinome, with and without known substrates, highlighting the applicability of the method. The sensitivity of this approach is evidenced by its ability to detect phosphorylation events from nanogram quantities of immunoprecipitated material, which allows for wider applicability of this method. To increase the information content of the quantitative kinase motifs, a sublibrary approach was used to expand the testable sequence space within a peptide library of approximately 100 members for CDK1, CDK7, and CDK9. Kinetic analysis of the HIV-1 Tat (transactivator of transcription)-positive transcription elongation factor b (P-TEFb) interaction allowed for localization of the P-TEFb phosphorylation site as well as characterization of the stimulatory effect of Tat on P-TEFb catalytic efficiency.


Subject(s)
Phosphopeptides/analysis , Protein Kinases/metabolism , Tandem Mass Spectrometry , Amino Acid Motifs , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase 9/metabolism , HIV-1/metabolism , Humans , Kinetics , Peptide Library , Phosphopeptides/chemistry , Phosphorylation , Positive Transcriptional Elongation Factor B/chemistry , Positive Transcriptional Elongation Factor B/metabolism , Substrate Specificity , tat Gene Products, Human Immunodeficiency Virus/metabolism
8.
J Lipid Res ; 57(2): 288-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26621918

ABSTRACT

The covalent attachment of a 14-carbon aliphatic tail on a glycine residue of nascent translated peptide chains is catalyzed in human cells by two N-myristoyltransferase (NMT) enzymes using the rare myristoyl-CoA (C(14)-CoA) molecule as fatty acid donor. Although, NMT enzymes can only transfer a myristate group, they lack specificity for C(14)-CoA and can also bind the far more abundant palmitoyl-CoA (C(16)-CoA) molecule. We determined that the acyl-CoA binding protein, acyl-CoA binding domain (ACBD)6, stimulated the NMT reaction of NMT2. This stimulatory effect required interaction between ACBD6 and NMT2, and was enhanced by binding of ACBD6 to its ligand, C(18:2)-CoA. ACBD6 also interacted with the second human NMT enzyme, NMT1. The presence of ACBD6 prevented competition of the NMT reaction by C(16)-CoA. Mutants of ACBD6 that were either deficient in ligand binding to the N-terminal ACBD or unable to interact with NMT2 did not stimulate activity of NMT2, nor could they protect the enzyme from utilizing the competitor C(16)-CoA. These results indicate that ACBD6 can locally sequester C(16)-CoA and prevent its access to the enzyme binding site via interaction with NMT2. Thus, the ligand binding properties of the NMT/ACBD6 complex can explain how the NMT reaction can proceed in the presence of the very abundant competitive substrate, C(16)-CoA.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Acyl Coenzyme A/metabolism , Acyltransferases/metabolism , Membrane Lipids/metabolism , Myristic Acid/metabolism , ATP-Binding Cassette Transporters/chemistry , Acylation , Acyltransferases/chemistry , Carrier Proteins , Coenzyme A/metabolism , Fatty Acids/genetics , Fatty Acids/metabolism , Humans , Membrane Lipids/chemistry , Palmitoyl Coenzyme A/metabolism , Phospholipids/metabolism , Protein Interaction Domains and Motifs/genetics , Substrate Specificity
9.
Nat Methods ; 9(11): 1095-100, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023596

ABSTRACT

We developed a simple and rapid multiplex substrate-profiling method to reveal the substrate specificity of any endo- or exopeptidase using liquid chromatography-tandem mass spectrometry sequencing. We generated a physicochemically diverse library of peptides by incorporating all combinations of neighbor and near-neighbor amino acid pairs into decapeptide sequences that are flanked by unique dipeptides at each terminus. Addition of a panel of evolutionarily diverse peptidases to a mixture of these tetradecapeptides generated information on prime and nonprime sites as well as on substrate specificity that matched or expanded upon known substrate motifs. This method biochemically confirmed the activity of the klassevirus 3C protein responsible for polypeptide processing and allowed granzyme B substrates to be ranked by enzymatic turnover efficiency using label-free quantitation of precursor-ion abundance. Additionally, the proteolytic secretions from schistosome parasitic flatworm larvae and a pancreatic cancer cell line were deconvoluted in a subtractive strategy using class-specific peptidase inhibitors.


Subject(s)
Peptide Hydrolases/metabolism , Substrate Specificity , 3C Viral Proteases , Animals , Carboxypeptidases/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cathepsin E/metabolism , Cell Line, Tumor , Chromatography, Liquid , Cysteine Endopeptidases/metabolism , Exopeptidases/metabolism , Granzymes/metabolism , Humans , Mice , Pancreatic Elastase/metabolism , Peptide Library , Peptides/metabolism , Schistosoma mansoni , Tandem Mass Spectrometry , Viral Proteins/metabolism
10.
J Biol Chem ; 288(18): 12489-99, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23504313

ABSTRACT

The transmembrane serine protease MarP is important for pH homeostasis in Mycobacterium tuberculosis (Mtb). Previous structural studies revealed that MarP contains a chymotrypsin fold and a disulfide bond that stabilizes the protease active site in the substrate-bound conformation. Here, we determined that MarP is located in the Mtb periplasm and showed that this localization is essential for function. Using the recombinant protease domain of MarP, we identified its substrate specificity using two independent assays: positional-scanning synthetic combinatorial library profiling and multiplex substrate profiling by mass spectrometry. These methods revealed that MarP prefers bulky residues at P4, tryptophan or leucine at P2, arginine or hydrophobic residues at P1, and alanine or asparagine at P1'. Guided by these data, we designed fluorogenic peptide substrates and characterized the kinetic properties of MarP. Finally, we tested the impact of mutating MarP cysteine residues on the peptidolytic activity of recombinant MarP and its ability to complement phenotypes of Mtb ΔMarP. Taken together, our studies provide insight into the enzymatic properties of MarP, its substrate preference, and the importance of its transmembrane helices and disulfide bond.


Subject(s)
Mycobacterium tuberculosis/enzymology , Oxidative Stress/physiology , Peptide Hydrolases/metabolism , Periplasmic Proteins/metabolism , Protein Folding , Hydrogen-Ion Concentration , Mutation , Mycobacterium tuberculosis/genetics , Peptide Hydrolases/genetics , Periplasmic Proteins/genetics , Protein Structure, Secondary , Substrate Specificity/physiology
11.
Nat Struct Mol Biol ; 31(4): 621-632, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38216659

ABSTRACT

Multidrug resistance protein 4 (MRP4) is a broadly expressed ATP-binding cassette transporter that is unique among the MRP subfamily for transporting prostanoids, a group of signaling molecules derived from unsaturated fatty acids. To better understand the basis of the substrate selectivity of MRP4, we used cryogenic-electron microscopy to determine six structures of nanodisc-reconstituted MRP4 at various stages throughout its transport cycle. Substrate-bound structures of MRP4 in complex with PGE1, PGE2 and the sulfonated-sterol DHEA-S reveal a common binding site that accommodates a diverse set of organic anions and suggest an allosteric mechanism for substrate-induced enhancement of MRP4 ATPase activity. Our structure of a catalytically compromised MRP4 mutant bound to ATP-Mg2+ is outward-occluded, a conformation previously unobserved in the MRP subfamily and consistent with an alternating-access transport mechanism. Our study provides insights into the endogenous function of this versatile efflux transporter and establishes a basis for MRP4-targeted drug design.


Subject(s)
Multidrug Resistance-Associated Proteins , Prostaglandins , Prostaglandins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Biological Transport , Dinoprostone/metabolism , Membrane Transport Proteins/metabolism
12.
J Biol Chem ; 287(25): 21152-63, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22539347

ABSTRACT

To identify the gut-associated tick aspartic hemoglobinase, this work focuses on the functional diversity of multiple Ixodes ricinus cathepsin D forms (IrCDs). Out of three encoding genes representing Ixodes scapularis genome paralogs, IrCD1 is the most distinct enzyme with a shortened propeptide region and a unique pattern of predicted post-translational modifications. IrCD1 gene transcription is induced by tick feeding and is restricted to the gut tissue. The hemoglobinolytic role of IrCD1 was further supported by immunolocalization of IrCD1 in the vesicles of tick gut cells. Properties of recombinantly expressed rIrCD1 are consistent with the endo-lysosomal environment because the zymogen is autoactivated and remains optimally active in acidic conditions. Hemoglobin cleavage pattern of rIrCD1 is identical to that produced by the native enzyme. The preference for hydrophobic residues at the P1 and P1' position was confirmed by screening a novel synthetic tetradecapeptidyl substrate library. Outside the S1-S1' regions, rIrCD1 tolerates most amino acids but displays a preference for tyrosine at P3 and alanine at P2'. Further analysis of the cleavage site location within the peptide substrate indicated that IrCD1 is a true endopeptidase. The role in hemoglobinolysis was verified with RNAi knockdown of IrCD1 that decreased gut extract cathepsin D activity by >90%. IrCD1 was newly characterized as a unique hemoglobinolytic cathepsin D contributing to the complex intestinal proteolytic network of mainly cysteine peptidases in ticks.


Subject(s)
Arthropod Proteins/metabolism , Cathepsin D/metabolism , Hemoglobins/metabolism , Intestines/enzymology , Ixodes/enzymology , Protein Processing, Post-Translational/physiology , Animals , Arthropod Proteins/genetics , Cathepsin D/genetics , Genome/physiology , Hemoglobins/genetics , Ixodes/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription, Genetic/physiology
13.
J Virol ; 86(7): 3605-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258260

ABSTRACT

The activity of phosphatidylinositol 4-kinase class III beta (PI4KIIIß) has been shown to be required for the replication of multiple picornaviruses; however, it is unclear whether a physical association between PI4KIIIß and the viral replication machinery exists and, if it does, whether association is necessary. We examined the ability of the 3A protein from 18 different picornaviruses to form a complex with PI4KIIIß by affinity purification of Strep-Tagged transiently transfected constructs followed by mass spectrometry and Western blotting for putative interacting targets. We found that the 3A proteins of Aichi virus, bovine kobuvirus, poliovirus, coxsackievirus B3, and human rhinovirus 14 all copurify with PI4KIIIß. Furthermore, we found that multiple picornavirus 3A proteins copurify with the Golgi adaptor protein acyl coenzyme A (acyl-CoA) binding domain protein 3 (ACBD3/GPC60), including those from Aichi virus, bovine kobuvirus, human rhinovirus 14, poliovirus, and coxsackievirus B2, B3, and B5. Affinity purification of ACBD3 confirmed interaction with multiple picornaviral 3A proteins and revealed the ability to bind PI4KIIIß in the absence of 3A. Mass-spectrometric analysis of transiently expressed Aichi virus, bovine kobuvirus, and human klassevirus 3A proteins demonstrated that the N-terminal glycines of these 3A proteins are myristoylated. Alanine-scanning mutagenesis along the entire length of Aichi virus 3A followed by transient expression and affinity purification revealed that copurification of PI4KIIIß could be eliminated by mutation of specific residues, with little or no effect on recruitment of ACBD3. One mutation at the N terminus, I5A, significantly reduced copurification of both ACBD3 and PI4KIIIß. The dependence of Aichi virus replication on the activity of PI4KIIIß was confirmed by both chemical and genetic inhibition. Knockdown of ACBD3 by small interfering RNA (siRNA) also prevented replication of both Aichi virus and poliovirus. Point mutations in 3A that eliminate PI4KIIIß association sensitized Aichi virus to PIK93, suggesting that disruption of the 3A/ACBD3/PI4KIIIß complex may represent a novel target for therapeutic intervention that would be complementary to the inhibition of the kinase activity itself.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Picornaviridae Infections/metabolism , Picornaviridae/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Cell Line , Golgi Apparatus/genetics , Humans , Kobuvirus/genetics , Kobuvirus/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Minor Histocompatibility Antigens , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Picornaviridae/chemistry , Picornaviridae/genetics , Picornaviridae Infections/enzymology , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protein Binding , Sequence Alignment , Viral Nonstructural Proteins/genetics
14.
J Nat Prod ; 76(4): 630-41, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23445522

ABSTRACT

Almiramide C is a marine natural product with low micromolar activity against Leishmania donovani, the causative agent of leishmaniasis. We have now shown that almiramide C is also active against the related parasite Trypanosoma brucei, the causative agent of human African trypanosomiasis. A series of activity-based probes have been synthesized to explore both the molecular target of this compound series in T. brucei lysates and site localization through epifluorescence microscopy. These target identification studies indicate that the almiramides likely perturb glycosomal function through disruption of membrane assembly machinery. Glycosomes, which are organelles specific to kinetoplastid parasites, house the first seven steps of glycolysis and have been shown to be essential for parasite survival in the bloodstream stage. There are currently no reported small-molecule disruptors of glycosome function, making the almiramides unique molecular probes for this understudied parasite-specific organelle. Additionally, examination of toxicity in an in vivo zebrafish model has shown that these compounds have little effect on organism development, even at high concentrations, and has uncovered a potential side effect through localization of fluorescent derivatives to zebrafish neuromast cells. Combined, these results further our understanding of the potential value of this lead series as development candidates against T. brucei.


Subject(s)
Biological Products/pharmacology , Chagas Disease/drug therapy , Lipopeptides/pharmacology , Trypanosoma brucei brucei/drug effects , Animals , Biological Products/chemistry , Glycolysis/physiology , Humans , Leishmania donovani/drug effects , Microbodies/metabolism , Microscopy, Fluorescence , Trypanosoma brucei brucei/metabolism , Zebrafish/physiology
15.
Mol Neurodegener ; 18(1): 29, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37131250

ABSTRACT

BACKGROUND: Autosomal dominant mutations in α-synuclein, TDP-43 and tau are thought to predispose to neurodegeneration by enhancing protein aggregation. While a subset of α-synuclein, TDP-43 and tau mutations has been shown to increase the structural propensity of these proteins toward self-association, rates of aggregation are also highly dependent on protein steady state concentrations, which are in large part regulated by their rates of lysosomal degradation. Previous studies have shown that lysosomal proteases operate precisely and not indiscriminately, cleaving their substrates at very specific linear amino acid sequences. With this knowledge, we hypothesized that certain coding mutations in α-synuclein, TDP-43 and tau may lead to increased protein steady state concentrations and eventual aggregation by an alternative mechanism, that is, through disrupting lysosomal protease cleavage recognition motifs and subsequently conferring protease resistance to these proteins. RESULTS: To test this possibility, we first generated comprehensive proteolysis maps containing all of the potential lysosomal protease cleavage sites for α-synuclein, TDP-43 and tau. In silico analyses of these maps indicated that certain mutations would diminish cathepsin cleavage, a prediction we confirmed utilizing in vitro protease assays. We then validated these findings in cell models and induced neurons, demonstrating that mutant forms of α-synuclein, TDP-43 and tau are degraded less efficiently than wild type despite being imported into lysosomes at similar rates. CONCLUSIONS: Together, this study provides evidence that pathogenic mutations in the N-terminal domain of α-synuclein (G51D, A53T), low complexity domain of TDP-43 (A315T, Q331K, M337V) and R1 and R2 domains of tau (K257T, N279K, S305N) directly impair their own lysosomal degradation, altering protein homeostasis and increasing cellular protein concentrations by extending the degradation half-lives of these proteins. These results also point to novel, shared, alternative mechanism by which different forms of neurodegeneration, including synucleinopathies, TDP-43 proteinopathies and tauopathies, may arise. Importantly, they also provide a roadmap for how the upregulation of particular lysosomal proteases could be targeted as potential therapeutics for human neurodegenerative disease.


Subject(s)
DNA-Binding Proteins , Neurodegenerative Diseases , alpha-Synuclein , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Half-Life , Lysosomes/metabolism , Mutation/genetics , Neurodegenerative Diseases/metabolism , Peptide Hydrolases/metabolism , tau Proteins/genetics , tau Proteins/metabolism
16.
J Biol Chem ; 285(41): 31120-9, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20675366

ABSTRACT

Proteases are a ubiquitous group of enzymes that play key roles in the life cycle of parasites, in the host-parasite relationship, and in the pathogenesis of parasitic diseases. Furthermore, proteases are druggable targets for the development of new anti-parasitic therapy. The subtilisin protease (SUB; Clan SB, family S8) of Leishmania donovani was cloned and found to possess a unique catalytic triad. This gene was then deleted by gene knock-out, which resulted in reduced ability by the parasite to undergo promastigote to amastigote differentiation in vitro. Electron microscopy of SUB knock-out amastigotes revealed abnormal membrane structures, retained flagella, and increased binucleation. SUB-deficient Leishmania displayed reduced virulence in both hamster and murine infection models. Histology of spleens from SUB knock-out-infected hamsters revealed the absence of psammoma body calcifications indicative of the granulomatous lesions that occur during Leishmania infection. To delineate the specific role of SUB in parasite physiology, two-dimensional gel electrophoresis was carried out on SUB(-/-) versus wild-type parasites. SUB knock-out parasites showed altered regulation of the terminal peroxidases of the trypanothione reductase system. Leishmania and other trypanosomatids lack glutathione reductase, and therefore rely on the novel trypanothione reductase system to detoxify reactive oxygen intermediates and to maintain redox homeostasis. The predominant tryparedoxin peroxidases were decreased in SUB(-/-) parasites, and higher molecular weight isoforms were present, indicating altered processing. In addition, knock-out parasites showed increased sensitivity to hydroperoxide. These data suggest that subtilisin is the maturase for tryparedoxin peroxidases and is necessary for full virulence.


Subject(s)
Leishmania donovani/enzymology , Leishmania donovani/pathogenicity , NADH, NADPH Oxidoreductases/metabolism , Protozoan Proteins/metabolism , Subtilisin/metabolism , Animals , Cricetinae , Disease Models, Animal , Female , Gene Knockdown Techniques , Hydrogen Peroxide/pharmacology , Leishmania donovani/genetics , Male , Mesocricetus , Mice , Mice, Inbred BALB C , NADH, NADPH Oxidoreductases/genetics , Oxidants/pharmacology , Protozoan Proteins/genetics , Subtilisin/genetics
17.
ACS Chem Biol ; 16(4): 712-723, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33765766

ABSTRACT

Differential expression of extracellular proteases and endogenous protease inhibitors has been associated with distinct molecular subtypes of breast cancer. However, due to the tight post-translational regulation of protease activity, protease expression-level data alone are not sufficient to understand the role of proteases in malignant transformation. Therefore, we hypothesized that global profiles of extracellular protease activity could more completely reflect differences observed at the transcriptional level in breast cancer and that subtype-associated protease activity may be leveraged to identify specific proteases that play a functional role in cancer signaling. Here, we used a global peptide library-based approach to profile the activities of proteases within distinct breast cancer subtypes. Analysis of 3651 total peptide cleavages from a panel of well-characterized breast cancer cell lines demonstrated differences in proteolytic signatures between cell lines. Cell line clustering based on protease cleavages within the peptide library expanded upon the expected classification derived from transcriptional profiling. An isogenic cell line model developed to further interrogate proteolysis in the HER2 subtype revealed a proteolytic signature consistent with activation of TGF-ß signaling. Specifically, we determined that a metalloprotease involved in TGF-ß signaling, BMP1, was upregulated at both the protein (2-fold, P = 0.001) and activity (P = 0.0599) levels. Inhibition of BMP1 and HER2 suppressed invasion of HER2-expressing cells by 35% (P < 0.0001), compared to 15% (P = 0.0086) observed in cells where only HER2 was inhibited. In summary, through global identification of extracellular proteolysis in breast cancer cell lines, we demonstrate subtype-specific differences in protease activity and elucidate proteolysis associated with HER2-mediated signaling.


Subject(s)
Breast Neoplasms/metabolism , Genes, erbB-2 , Peptide Hydrolases/metabolism , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Humans , Proteolysis
18.
Neurology ; 93(5): e433-e444, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31270218

ABSTRACT

OBJECTIVE: To identify molecular correlates of primary angiitis of the CNS (PACNS) through proteomic analysis of CSF from a biopsy-proven patient cohort. METHODS: Using mass spectrometry, we quantitatively compared the CSF proteome of patients with biopsy-proven PACNS (n = 8) to CSF from individuals with noninflammatory conditions (n = 11). Significantly enriched molecular pathways were identified with a gene ontology workflow, and high confidence hits within enriched pathways (fold change >1.5 and concordant Benjamini-Hochberg-adjusted p < 0.05 on DeSeq and t test) were identified as differentially regulated proteins. RESULTS: Compared to noninflammatory controls, 283 proteins were differentially expressed in the CSF of patients with PACNS, with significant enrichment of the complement cascade pathway (C4-binding protein, CD55, CD59, properdin, complement C5, complement C8, and complement C9) and neural cell adhesion molecules. A subset of clinically relevant findings were validated by Western blot and commercial ELISA. CONCLUSIONS: In this exploratory study, we found evidence of deregulation of the alternative complement cascade in CSF from biopsy-proven PACNS compared to noninflammatory controls. More specifically, several regulators of the C3 and C5 convertases and components of the terminal cascade were significantly altered. These preliminary findings shed light on a previously unappreciated similarity between PACNS and systemic vasculitides, especially anti-neutrophil cytoplasmic antibody-associated vasculitis. The therapeutic implications of this common biology and the diagnostic and therapeutic utility of individual proteomic findings warrant validation in larger cohorts.


Subject(s)
Complement System Proteins/cerebrospinal fluid , Neural Cell Adhesion Molecules/cerebrospinal fluid , Proteomics , Vasculitis, Central Nervous System/cerebrospinal fluid , Adolescent , Adult , Biopsy , Brain/pathology , CD55 Antigens/cerebrospinal fluid , CD59 Antigens/cerebrospinal fluid , Case-Control Studies , Cohort Studies , Complement C4b-Binding Protein/cerebrospinal fluid , Complement C5/cerebrospinal fluid , Complement C8/cerebrospinal fluid , Complement C9/cerebrospinal fluid , Complement Pathway, Alternative , Female , Gene Ontology , Humans , Male , Mass Spectrometry , Middle Aged , Properdin/cerebrospinal fluid , Vasculitis, Central Nervous System/pathology
19.
J Am Chem Soc ; 130(30): 9624-5, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18593116

ABSTRACT

Accurate quantification of protein content and composition has been achieved using isotope-edited surface enhanced resonance Raman spectroscopy. Synthesis of isotopomeric Rhodamine dye-linked bioconjugation reagents enabled direct labeling of surface lysines on a variety of proteins. When separated in polyacrylamide gels and stained with silver nanoparticles. The spectral signatures reflect the expected statistical distribution of isotopomeric labels on the labeled proteins in the gel matrix format without interference from protein features.


Subject(s)
Fluorescent Dyes/chemistry , Proteins/analysis , Rhodamines/chemistry , Spectrum Analysis, Raman/methods , Isotope Labeling
20.
Biochimie ; 90(2): 345-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17936488

ABSTRACT

Schistosomes are parasitic platyhelminths (flatworms) of birds and mammals. As a parasitic disease of humans, schistosomiasis ranks second only to malaria in global importance. Schistosome larvae (cercariae) must invade and penetrate skin as an initial step to successful infection of the vertebrate host. Proteolytic enzymes secreted from the acetabular glands of cercariae contribute significantly to the invasion process. In this comparative study, we analyzed protease activities secreted by cercariae of Schistosoma mansoni, Schistosoma japonicum and Schistosomatium douthitti. Using protease-family specific, irreversible active-site probes, fluorogenic peptidyl substrates, immuno-histochemistry and high-resolution mass spectrometry, considerable species differences were noted in the quantity and character of proteases. Serine proteases, the most abundant enzymes secreted by S. mansoni cercariae, were not identified in S. japonicum. In contrast, the acetabular gland contents of S. japonicum cercariae had a 40-fold greater cathepsin B-like activity than those of S. mansoni. Based on the present data and previous reports, we propose that cysteine proteases represent an archetypal tool for tissue invasion among primitive metazoa and the use of serine proteases arose later in schistosome evolution. Computational analysis of serine protease phylogeny revealed an extraordinarily distant relationship between S. mansoni serine proteases and other members of the Clan PA family S1 proteases.


Subject(s)
Cathepsin B/metabolism , Schistosoma japonicum/enzymology , Schistosoma mansoni/enzymology , Serine Endopeptidases/metabolism , Animals , Cathepsin B/chemistry , Chymotrypsin/metabolism , Gene Transfer, Horizontal , Larva/enzymology , Larva/pathogenicity , Mass Spectrometry , Phylogeny , Proteomics , Schistosoma japonicum/growth & development , Schistosoma japonicum/pathogenicity , Schistosomatidae/enzymology , Serine Endopeptidases/classification , Serine Endopeptidases/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL