Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Arch Oral Biol ; 158: 105869, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38104461

ABSTRACT

OBJECTIVES: describing the clinical features of twelve Egyptian patients with Papillon-Lefever syndrome (PLS). Five novel mutations in the cathepsin C (CTSC) gene are introduced and the phenotype of the syndrome is expanded by the identification of new clinical features. DESIGN: the clinical, oro-dental data of twelve Egyptian patients from seven unrelated families are described. Sequence analysis of the CTSC gene was performed to identify the causative mutaions. RESULTS: Typical PLS features were presented in all patints but with variable severity. One patient showed atypical dental features including dental structural defect, minimal periodontitis, severe gingivitis, and delayed closure of root apices. Another patient presented with arachnodactyly, dystrophic nails, and buphthalmos in the right eye secondary to uncontrolled congenital glaucoma. Mutational analysis of CTSC gene revealed seven distinct homozygous variants including five novel ones: c.285_286delGT (p.Leu96GlufsTer2), c .302 G>C (p.Trp101Ser), c.622_628delCACAGTC (p.H208Efs*11), c.1331delinsAAAAA (p.G444Efs*4) and c .1343 G>A (p.Cys448Tyr). The previously reported missense variant c .757 G>A (p.Ala253Thr) was found in one patient. This variant is very close to the splice region and by functional studies, we proved that it results in exon skipping and early protein truncation (p.R214Sfs*46). CONCLUSION: We report five novel CTSC variants and describe rare and unusual associated clinical and dental findings such as dental structural defects, delayed closure of root apices, and congenital glaucoma. Therefore, our results expand both the phenotypic and mutational spectrum of PLS.


Subject(s)
Glaucoma , Papillon-Lefevre Disease , Humans , Papillon-Lefevre Disease/genetics , Cathepsin C/chemistry , Cathepsin C/genetics , Egypt , Mutation, Missense , Syndrome
2.
Int J Mol Sci ; 23(3)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35163774

ABSTRACT

Human dipeptidyl peptidase I (DPPI) belongs to the family of papain-like cysteine peptidases. Its distinctive features are the unique exclusion domain which enables the eponymous activity and homotetramerization of DPPI, and its dependence on chloride ions for enzymatic activity. The oligomeric state of DPPI is unique in this family of predominantly monomeric peptidases. However, a distant DPPI ortholog from Plasmodium falciparum has been shown to be monomeric, indicating that the oligomeric state of DPPI varies between lineages. The aim of this work was to study the evolution of DPPI, with particular attention to the structural features that determine its characteristic enzymatic activity and preferences, and to reconstruct the evolution of its oligomerization. We analyzed fifty-seven selected sequences of DPPI and confirmed its presence in three lineages, namely, Amorphea (including animals and Amoebozoa), Alveolates and the metamonad Giardia. The amino acid residues that bind the chloride ion are highly conserved in all species, indicating that the dependence on chloride ions for activity is an evolutionarily conserved feature of DPPI. The number of N-glycosylation sites is significantly increased in animals, particularly vertebrates. Analysis of homology models and subunit contacts suggests that oligomerization is likely restricted to DPPIs in the Amorphea group.


Subject(s)
Cathepsin C/chemistry , Cathepsin C/genetics , Alveolata/enzymology , Amoebozoa/enzymology , Evolution, Molecular , Giardia/enzymology , Glycosylation , Humans , Models, Molecular , Phylogeny , Protein Conformation , Protein Multimerization , Structural Homology, Protein
3.
Biochem Pharmacol ; 194: 114803, 2021 12.
Article in English | MEDLINE | ID: mdl-34678221

ABSTRACT

Epidemiological studies established an association between chronic inflammation and higher risk of cancer. Inhibition of proteolytic enzymes represents a potential treatment strategy for cancer and prevention of cancer metastasis. Cathepsin C (CatC) is a highly conserved lysosomal cysteine dipeptidyl aminopeptidase required for the activation of pro-inflammatory neutrophil serine proteases (NSPs, elastase, proteinase 3, cathepsin G and NSP-4). NSPs are locally released by activated neutrophils in response to pathogens and non-infectious danger signals. Activated neutrophils also release neutrophil extracellular traps (NETs) that are decorated with several neutrophil proteins, including NSPs. NSPs are not only NETs constituents but also play a role in NET formation and release. Although immune cells harbor large amounts of CatC, additional cell sources for this protease exists. Upregulation of CatC expression was observed in different tissues during carcinogenesis and correlated with metastasis and poor patient survival. Recent mechanistic studies indicated an important interaction of tumor-associated CatC, NSPs, and NETs in cancer development and metastasis and suggested CatC as a therapeutic target in a several cancer types. Cancer cell-derived CatC promotes neutrophil recruitment in the inflammatory tumor microenvironment. Because the clinical consequences of genetic CatC deficiency in humans resulting in the elimination of NSPs are mild, small molecule inhibitors of CatC are assumed as safe drugs to reduce the NSP burden. Brensocatib, a nitrile CatC inhibitor is currently tested in a phase 3 clinical trial as a novel anti-inflammatory therapy for patients with bronchiectasis. However, recently developed CatC inhibitors possibly have protective effects beyond inflammation. In this review, we describe the pathophysiological function of CatC and discuss molecular mechanisms substantiating pharmacological CatC inhibition as a potential strategy for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cathepsin C/antagonists & inhibitors , Cathepsin C/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Cathepsin C/chemistry , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Serine Proteases/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
4.
Arch Biochem Biophys ; 675: 108121, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31574257

ABSTRACT

Human dipeptidyl-peptidase I (DPPI) is a tetrameric enzyme from the family of papain-like cysteine peptidases. It is ubiquitously expressed and plays important roles in general protein turnover, skin homeostasis and proteolytic processing of effector peptidases in immune cells. In this work we investigate allosteric regulation of DPPI and its relation to the oligomeric structure. First, we investigate the functional significance of the tetrameric state by comparing the kinetic properties of the tetrameric form (DPPItet) with a recombinant monomeric form (DPPImono). We find that both forms have very similar kinetic properties for the hydrolysis of a commonly used synthetic substrate. In agreement with previous studies, no cooperativity is observed in the tetramer. The only significant difference between both forms is a higher catalytic rate of DPPImono. We then characterize three compounds, 3'-nitrophthalanilic acid, chlorogenic acid and caffeic acid that affect DPPI activity via kinetic mechanisms consistent with binding outside of the active site. These compounds are the first known modifiers of DPPI that do not act as specific inhibitors. Chlorogenic acid and caffeic acid act as linear mixed and linear catalytic inhibitors, respectively, and do not discriminate between both forms. In contrast, 3'-nitrophthalanilic acid is a hyperbolic inhibitor that binds DPPItet and DPPImono with different affinities and inhibits their activities via different kinetic mechanisms. Altogether, these results show that the tetrameric structure of DPPI is not necessary for enzymatic activity, however, oligomerization-related structural features can play a role in its regulation.


Subject(s)
Cathepsin C/metabolism , Allosteric Regulation , Cathepsin C/chemistry , Humans , Hydrolysis , Kinetics , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
5.
Int J Mol Sci ; 20(19)2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31557781

ABSTRACT

Cysteine cathepsin C (CatC) is a ubiquitously expressed, lysosomal aminopeptidase involved in the activation of zymogens of immune-cell-associated serine proteinases (elastase, cathepsin G, proteinase 3, neutrophil serine proteinase 4, lymphocyte granzymes, and mast cell chymases). CatC is first synthetized as an inactive zymogen containing an intramolecular chain propeptide, the dimeric form of which is processed into the mature tetrameric form by proteolytic cleavages. A molecular modeling analysis of proCatC indicated that its propeptide displayed a similar fold to those of other lysosomal cysteine cathepsins, and could be involved in dimer formation. Our in vitro experiments revealed that human proCatC was processed and activated by CatF, CatK, and CatV in two consecutive steps of maturation, as reported for CatL and CatS previously. The unique positioning of the propeptide domains in the proCatC dimer complex allows this order of cleavages to be understood. The missense mutation Leu172Pro within the propeptide region associated with the Papillon-Lefèvre and Haim-Munk syndrome altered the proform stability as well as the maturation of the recombinant Leu172Pro proform.


Subject(s)
Cathepsin C/chemistry , Enzyme Precursors/chemistry , Models, Molecular , Molecular Conformation , Binding Sites , Humans , Protein Binding , Recombinant Proteins/chemistry
6.
Am J Med Genet A ; 179(10): 2124-2131, 2019 10.
Article in English | MEDLINE | ID: mdl-31282082

ABSTRACT

Papillon-Lefèvre syndrome (PLS; MIM#245000) is a rare recessive autosomal disorder characterized by palmar and plantar hyperkeratosis, and aggressively progressing periodontitis leading to premature loss of deciduous and permanent teeth. PLS is caused by loss-of-function mutations in the CTSC gene, which encodes cathepsin C. PLS clinical expressivity is highly variable and no consistent genotype-phenotype correlation has been demonstrated yet. Here we report the clinical and genetic features of five PLS patients presenting a severe periodontal breakdown in primary and permanent dentition, hyperkeratosis over palms and soles, and recurrent sinusitis and/or tonsillitis. Mutation analysis revealed two novel homozygous recessive mutations (c.947T>C and c.1010G>C) and one previous described homozygous recessive mutation (c.901G>A), with parents carrying them in heterozygous, in three families (four patients). The fourth family presented with the CTSC c.628C>T mutation in heterozygous, which was inherited maternally. Patient carrying the CTSC c.628C>T mutation featured classical PLS phenotype, but no PLS clinical characteristics were found in his carrier mother. All mutations were found to affect directly (c.901G>A, c.947T>C, and c.1010G>C) or indirectly (c.628C>T, which induces a premature termination) the heavy chain of the cathepsin C, the region responsible for activation of the lysosomal protease. Together, these findings indicate that both homozygous and heterozygous mutations in the cathepsin C heavy chain domain may lead to classical PLS phenotype, suggesting roles for epistasis or gene-environment interactions on determination of PLS phenotypes.


Subject(s)
Papillon-Lefevre Disease/genetics , Papillon-Lefevre Disease/pathology , Adolescent , Adult , Cathepsin C/chemistry , Child , Child, Preschool , Female , Humans , Male , Models, Molecular , Papillon-Lefevre Disease/diagnostic imaging , Young Adult
7.
J Med Chem ; 62(12): 5901-5919, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31145622

ABSTRACT

In the course of developing the biochemistry to chemistry activity-based protein profiling (BTC-ABPP) method, we herein unexpectedly discovered that the epidermal growth factor receptor irreversible inhibitor WZ4002 also functioned as a low micromolar inhibitor of cathepsin C (CatC), a promising target for the treatment of numerous inflammatory and autoimmune diseases. Building on from this discovery, and following structure-activity relationship investigations guided by computational modeling, a novel series of pyridine scaffold compounds were developed as irreversible CatC inhibitors, further culminated in identifying a highly potent and selective inhibitor 22, which displays good metabolic stability and oral bioavailability. In vivo studies revealed that compound 22 clearly displays the ability to inhibit CatC, consequently leading to efficient inhibition of downstream neutrophil serine proteases in both bone marrow and blood. The overall excellent profile of compound 22 made it an interesting candidate for further preclinical investigation.


Subject(s)
Acrylamides/chemistry , Acrylamides/pharmacology , Cathepsin C/antagonists & inhibitors , Drug Design , ErbB Receptors/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Acrylamides/metabolism , Acrylamides/pharmacokinetics , Animals , Catalytic Domain , Cathepsin C/chemistry , Cathepsin C/metabolism , Cell Line , Humans , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics
8.
J Hum Genet ; 64(7): 689-694, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31068678

ABSTRACT

Cathepsin C (CatC) is a cysteine protease involved in a variety of immune and inflammatory pathways such as activation of cytotoxicity of various immune cells. Homozygous or compound heterozygous variants in the CatC coding gene CTSC cause different conditions that have in common severe periodontitis. Periodontitis may occur as part of Papillon-Lefèvre syndrome (PLS; OMIM#245000) or Haim-Munk syndrome (HMS; OMIM#245010), or may present as an isolated finding named aggressive periodontitis (AP1; OMIM#170650). AP1 generally affects young children and results in destruction of the periodontal support of the primary dentition. In the present study we report exome sequencing of a three generation consanguineous Turkish family with a recessive form of early-onset AP1. We identified a novel homozygous missense variant in exon 2 of CTSC (NM_148170, c.G302C, p.Trp101Ser) predicted to disrupt protein structure and to be disease causing. This is the first described CTSC variant specific to the nonsyndromic AP1 form. Given the broad phenotypic spectrum associated with CTSC variants, reporting this novel variant gives new insights on genotype/phenotype correlations and might improve diagnosis of patients with early-onset AP1.


Subject(s)
Aggressive Periodontitis/genetics , Cathepsin C/genetics , Mutation, Missense , Adolescent , Adult , Aggressive Periodontitis/diagnostic imaging , Cathepsin C/chemistry , Child , Female , Genetic Association Studies , Homozygote , Humans , Male , Turkey , Exome Sequencing
9.
Oral Dis ; 25(5): 1394-1402, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30908832

ABSTRACT

OBJECTIVES: To identify the molecular basis of Papillon-Lefèvre syndrome in two Chinese families. METHODS: Peripheral blood and mouth swab samples were obtained, from which genomic DNA and RNA were isolated. Sanger sequencing was employed to identify the mutations. mRNA expression was tested by real-time quantitative PCR. Evolutionary conservation, pathogenicity prediction and impact of protein structures of the mutations were conducted with bioinformatics tools and homology modelling. HEK293 cells were transfected with plasmids expressing wild-type or mutated CTSC. CTSC protein expression level and enzyme activity were explored. RESULTS: Mutation analysis revealed two novel compound heterozygous mutations, the c.190-191insA and c.1211-1212delA in patient 1 and the c.716A>G and c.757+1G>A in patient 2. In both patients, the levels of CTSC mRNA were significantly lower than in their relatives. Homology modelling analysis predicted that the mutations affect the structure and stability of the protein, and in vitro study showed that the CTSC proteins containing the mutations c.190-191insA and c.1211-1212delA, which result in truncated versions of protein, display impaired enzyme activity. The protein containing c.716A>G mutation showed quite similar enzyme activity compared to wild-type CTSC. CONCLUSION: Our data support the molecular mechanism of PLS and enlarge the scope of CTSC gene mutations related to PLS.


Subject(s)
Cathepsin C/genetics , Papillon-Lefevre Disease/complications , Amino Acid Sequence , Cathepsin C/chemistry , Cathepsin C/metabolism , DNA Mutational Analysis , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Papillon-Lefevre Disease/diagnosis , Papillon-Lefevre Disease/genetics
10.
Protein Expr Purif ; 157: 21-27, 2019 05.
Article in English | MEDLINE | ID: mdl-30703555

ABSTRACT

Cathepsin C is a tetrameric lysosomal protease that acts as a dipeptidyl-peptidase due to the presence of the exclusion domain that is unique among papain-like cysteine proteases. Here we describe a recombinant form of cathepsin C lacking its exclusion domain (CatCΔEx) produced in a bacterial expression system (E. coli). CatCΔEx is a monomer with endoprotease activity and affinity for hydrophobic residues such as Phe, Leu or Pro, but not Val, in the P2 position. As opposed to cathepsin C, it does not require chloride ions for its activity. Despite lower turnover rates of hydrolysis of synthetic substrates, CatCΔEx has elastolytic and gelatinolytic activity comparable to other cysteine cathepsins.


Subject(s)
Cathepsin C/metabolism , Animals , Catalytic Domain , Cathepsin C/chemistry , Cathepsin C/genetics , Cattle , Collagen/metabolism , Elastin/metabolism , Enzyme Activation , Escherichia coli/genetics , Gelatin/metabolism , Humans , Kinetics , Models, Molecular , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
11.
Fish Shellfish Immunol ; 84: 423-433, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30308297

ABSTRACT

The lysosomal cysteine protease cathepsin C plays a pivotal role in regulation of inflammatory and immune responses. However, the function of fish cathepsin C in virus replication remains largely unknown. In this study, cathepsin C gene (Ec-CC) was cloned and characterized from orange-spotted grouper, Epinephelus coioides. The full-length Ec-CC cDNA was composed of 2077 bp. It contained an open reading frame (ORF) of 1374 bp and encoded a 458-amino acid protein which shared 89% identity to cathepsin C from bicolor damselfish (Stegastes partitus). Amino acid alignment analysis showed that Ec-CC contained an N-terminal signal peptide, the propeptide region and the mature peptide. RT-PCR analysis showed that Ec-CC transcript was expressed in all the examined tissues which abundant in spleen and head kidney. After challenged with Singapore grouper iridovirus (SGIV) stimulation, the relative expression of EC-CC was significantly increased at 24 h post-infection. Subcellular localization analysis revealed that Ec-CC was distributed mainly in the cytoplasm. Further studies showed that overexpression of Ec-CC in vitro significantly delayed the cytopathic effect (CPE) progression evoked by SGIV and inhibited the viral genes transcription. Moreover, overexpression of Ec-CC significantly increased the expression of proinflammatory cytokines during SGIV infection. Taken together, our results demonstrated that Ec-CC might play a functional role in SGIV infection by regulating the inflammation response.


Subject(s)
Bass/genetics , Bass/immunology , Cathepsin C/genetics , Cathepsin C/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Base Sequence , Cathepsin C/chemistry , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Hemorrhagic Disease Virus, Epizootic/physiology , Phylogeny , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Sequence Alignment/veterinary
12.
Arch Biochem Biophys ; 612: 91-102, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27746119

ABSTRACT

Cathepsin C is a widely expressed cysteine exopeptidase that is mostly recognized for the activation of the granule-associated proinflammatory serine proteases in neutrophils, cytotoxic T lymphocytes and mast cells. It has been shown that the enzyme can be secreted extracellularly; however, its occurrence in human bodily fluids/physiological samples has not been thoroughly studied. In the course of this study, the first fluorescence resonance energy transfer peptides for the measurement of the activity of human cathepsin C were designed and synthesized. Two series of tetra- and pentapeptide substrates enabled the detailed S' specificity study of cathepsin C, which has been examined for the first time. The extensive enzymatic studies of the obtained compounds resulted in the selection of the highly specific and selective substrate Thi-Ala(Mca)-Ser-Gly-Tyr(3-NO2)-NH2, which was successfully employed for the detection of cathepsin C activity in complex biological samples such as cell lysates, urine and bronchoalveolar lavage fluids. Molecular docking of the selected substrate was performed in order to better understand the binding mode of the substrates in the active site of cathepsin C.


Subject(s)
Cathepsin C/chemistry , Microscopy, Fluorescence/methods , Catalytic Domain , Cathepsin L/chemistry , Fluorescence Resonance Energy Transfer , Humans , Hydrogen-Ion Concentration , Inflammation , Kinetics , Mast Cells/cytology , Molecular Conformation , Molecular Docking Simulation , Neutrophils/metabolism , Peptides/chemistry , Protein Binding , Recombinant Proteins/chemistry , Substrate Specificity , T-Lymphocytes, Cytotoxic/cytology
13.
Methods Enzymol ; 559: 71-97, 2015.
Article in English | MEDLINE | ID: mdl-26096504

ABSTRACT

Here, we present protocols describing the use of the dipeptidyl-aminopeptidase-1 (DPP1, DAPase) exoprotease-based TAGZyme system and the endoprotease, Factor Xa. Both enable the recovery of proteins free of any amino acids encoded by the vector and/or protease recognition site. They also provide the possibility of removing the proteases from the preparation of the target protein by a simple subtractive chromatography step. TAGZyme enzymes contain an uncleavable His tag for removal by Immobilized Metal Ion Affinity Chromatography (IMAC). Factor Xa can be removed using Xa Removal Resin.


Subject(s)
Chromatography, Affinity/methods , Exopeptidases/chemistry , Animals , Biological Products/chemistry , Buffers , Cathepsin C/chemistry , Chromatography, Affinity/instrumentation , Factor Xa/chemistry , Glutathione Transferase/chemistry , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Ions , Metals/chemistry , Proteolysis , Recombinant Proteins/chemistry
14.
Parasitol Res ; 113(10): 3621-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25138068

ABSTRACT

Cathepsin C is an important exopeptidase of papain superfamily and plays a number of great important roles during the parasitic life cycle. The amino acid sequence of cathepsin C from Clonorchis sinensis (C. sinensis) showed 54, 53, and 49% identities to that of Schistosoma japonicum, Schistosoma mansoni, and Homo sapiens, respectively. Phylogenetic analysis utilizing the sequences of papain superfamily of C. sinensis demonstrated that cathepsin C and cathepsin Bs came from a common ancestry. Cathepsin C of C. sinensis (Cscathepsin C) was identified as an excretory/secretory product by Western blot analysis. The results of transcriptional level and translational level of Cscathepsin C at metacercaria stage were higher than that at adult worms. Immunolocalization analysis indicated that Cscathepsin C was specifically distributed in the suckers (oral sucker and ventral sucker), eggs, vitellarium, intestines, and testis of adult worms. In the metacercaria, it was mainly detected on the cyst wall and excretory bladder. Combining with the results mentioned above, it implies that Cscathepsin C may be an essential proteolytic enzyme for proteins digestion of hosts, nutrition assimilation, and immune invasion of C. sinensis. Furthermore, it may be a potential diagnostic antigen and drug target against C. sinensis infection.


Subject(s)
Cathepsin C/genetics , Clonorchiasis/parasitology , Clonorchis sinensis/enzymology , Amino Acid Motifs , Amino Acid Sequence , Animals , Antibodies, Helminth/immunology , Cathepsin C/chemistry , Cathepsin C/metabolism , Cats , Cloning, Molecular , Clonorchis sinensis/genetics , Clonorchis sinensis/immunology , Computational Biology , Cyprinidae/parasitology , Exopeptidases/chemistry , Exopeptidases/genetics , Exopeptidases/metabolism , Female , Humans , Immunohistochemistry , Male , Metacercariae , Models, Structural , Molecular Sequence Data , Papain/chemistry , Papain/genetics , Papain/metabolism , Phylogeny , Rats , Sequence Alignment
15.
Amino Acids ; 46(4): 931-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24381006

ABSTRACT

Mammalian cathepsin C is primarily responsible for the removal of N-terminal dipeptides and activation of several serine proteases in inflammatory or immune cells, while its malarial parasite ortholog dipeptidyl aminopeptidase 1 plays a crucial role in catabolizing the hemoglobin of its host erythrocyte. In this report, we describe the systematic substrate specificity analysis of three cathepsin C orthologs from Homo sapiens (human), Bos taurus (bovine) and Plasmodium falciparum (malaria parasite). Here, we present a new approach with a tailored fluorogenic substrate library designed and synthesized to probe the S1 and S2 pocket preferences of these enzymes with both natural and a broad range of unnatural amino acids. Our approach identified very efficiently hydrolyzed substrates containing unnatural amino acids, which resulted in the design of significantly better substrates than those previously known. Additionally, in this study significant differences in terms of the structures of optimal substrates for human and malarial orthologs are important from the therapeutic point of view. These data can be also used for the design of specific inhibitors or activity-based probes.


Subject(s)
Amino Acids/chemistry , Cathepsin C/chemistry , Dipeptides/chemistry , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Amino Acids/metabolism , Animals , Cathepsin C/metabolism , Cattle , Dipeptides/chemical synthesis , Dipeptides/metabolism , Humans , Kinetics , Molecular Structure , Plasmodium falciparum/chemistry , Protozoan Proteins/metabolism , Substrate Specificity
16.
Gene ; 538(1): 182-7, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24374475

ABSTRACT

BACKGROUND: Papillon-Lefèvre syndrome (PLS) is a rare autosomal recessive disorder characterized by hyperkeratosis involving the palms, soles, elbows, and knees followed by periodontitis, destruction of alveolar bone, and loss of primary and permanent teeth. Mutations of the lysosomal protease cathepsin C gene (CTSC) have been shown to be the genetic cause of PLS. This study analyzed CTSC mutations in five Iranian families with PLS and modeled the protein for mutations found in two of them. METHODS: DNA analysis was performed by direct automated sequencing of genomic DNA amplified from exonic regions and associated splice intron site junctions of CTSC. RFLP analyses were performed to investigate the presence of previously unidentified mutation(s) in control groups. Protein homology modeling of the deduced novel mutations (P35 delL and R272P) was performed using the online Swiss-Prot server for automated modeling and analyzed and tested with special bioinformatics tools to better understand the structural effects caused by mutations in cathepsin C protein (CTSC). RESULTS: Six Iranian patients with PLS experienced premature tooth loss and palm plantar hyperkeratosis. Sequence analysis of CTSC revealed a novel mutation (P35delL) in exon 1 of Patient 1, and four previously reported mutations; R210X in Patient 2, R272P in Patient 3, Q312R in two siblings of family 4 (Patients 4 and 5), and CS043636 in Patient 6. RFLP analyses revealed different restriction fragment patterns between 50 healthy controls and patients for the P35delL mutation. Modeling of the mutations found in CTSC, P35delL in Patient 1 and R272P in Patient 3 revealed structural effects, which caused the functional abnormalities of the mutated proteins. CONCLUSIONS: The presence of this mutation in these patients provides evidence for founder CTSC mutations in PLS. This newly identified P35delL mutation leads to the loss of a leucine residue in the protein. The result of this study indicates that the phenotypes observed in these two patients are likely due to CTSC mutations. Also, structural analyses of the altered proteins identified changes in energy and stereochemistry that likely alter protein function.


Subject(s)
Cathepsin C/genetics , Models, Molecular , Mutation , Papillon-Lefevre Disease/genetics , Adolescent , Amino Acid Sequence , Case-Control Studies , Cathepsin C/chemistry , Child , Female , Humans , Male , Molecular Sequence Data , Papillon-Lefevre Disease/diagnosis , Protein Conformation , Young Adult
17.
Fish Shellfish Immunol ; 33(4): 821-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22885030

ABSTRACT

Cathepsin C (Cath C) is a lysosomal cysteine protease that belongs to the papain superfamily. Cath C is capable of activating many chymotrypsin-like serine proteases and is reported to be a central coordinator for the activation of many serine proteinases in immune and inflammatory cells. In this study, Cath C cDNA was cloned from Fenneropenaeus chinensis (Fc). The complete cDNA of Fc-Cath C in Chinese white shrimp was found to be 1445-base pairs (bp) long. It contained an open reading frame (ORF) 1356-bp long and encoded a 451-amino acid residue protein, including a 17-amino acid residue signal peptide. Real-time PCR analysis results indicated that Fc-Cath C was present in all the tissues detected and exhibited high level of transcription in the hepatopancreas. In hemocytes, hepatopancreas, gills and intestine, Fc-Cath C was upregulated after stimulation by the Vibrio anguillarum and the white spot syndrome viruses (WSSVs). Replication of the WSSV increased after the injection of Fc-Cath C antiserum or knockdown Cath C by RNA interference. These results implied that Cath C might play a crucial role in the antiviral immune response of shrimp.


Subject(s)
Arthropod Proteins/immunology , Cathepsin C/immunology , Penaeidae/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Base Sequence , Cathepsin C/chemistry , Cathepsin C/genetics , Cathepsin C/metabolism , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression Profiling/veterinary , Gene Expression Regulation , Immunity, Innate , Injections, Intraperitoneal/veterinary , Molecular Sequence Data , Organ Specificity , Penaeidae/genetics , Penaeidae/virology , Phylogeny , Real-Time Polymerase Chain Reaction/veterinary , Recombinant Proteins/metabolism , Sequence Alignment/veterinary , Vibrio/metabolism , White spot syndrome virus 1/immunology
18.
Biochemistry ; 51(38): 7551-68, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22928782

ABSTRACT

We examined the cathepsin C-catalyzed hydrolysis of dipeptide substrates of the form Yaa-Xaa-AMC, using steady-state and pre-steady-state kinetic methods. The substrates group into three kinetic profiles based upon the broad range observed for k(cat)/K(a) and k(cat) values, pre-steady-state time courses, and solvent kinetic isotope effects (sKIEs). The dipeptide substrate Gly-Arg-AMC displayed large values for k(cat)/K(a) (1.6 ± 0.09 µM(-1) s(-1)) and k(cat) (255 ± 6 s(-1)), an inverse sKIE on k(cat)/K(a) ((D)(k(cat)/K(a)) = 0.6 ± 0.15), a modest, normal sKIE on k(cat) ((D)k(cat) = 1.6 ± 0.2), and immeasurable pre-steady-state kinetics, indicating an extremely fast pre-steady-state rate (>400 s(-1)). (Errors on fitted values are omitted in the text for clarity but may be found in Table 2.) These results conformed to a kinetic model where the acylation (k(ac)) and deacylation (k(dac)) half-reactions are very fast and similar in value. The second substrate type, Gly-Tyr-AMC and Ser-Tyr-AMC, the latter the subject of a comprehensive kinetic study (Schneck et al. (2008) Biochemistry 47, 8697-8710), were found to be less active substrates compared to Gly-Arg-AMC, with respective k(cat)/K(a) values of 0.49 ± 0.07 µM(-1 )s(-1) and 5.3 ± 0.5 µM(-1 )s(-1), and k(cat) values of 28 ± 1 s(-1) and 25 ± 0.5 s(-1). Solvent kinetic isotope effects for Ser-Tyr-AMC were found to be inverse for k(cat)/K(a) ((D)(k(cat)/K(a)) = 0.74 ± 0.05) and normal for k(cat) ((D)k(cat) = 2.3 ± 0.1) but unlike Gly-Arg-AMC, pre-steady-state kinetics of Gly-Tyr-AMC and Ser-Tyr-AMC were measurable and characterized by a single-exponential burst, with fast transient rates (490 s(-1) and 390 s(-1), respectively), from which it was determined that k(ac) ≫ k(dac) ∼ k(cat). The third substrate type, Gly-Ile-AMC, gave very low values of k(cat)/K(a) (0.0015 ± 0.0001 µM(-1) s(-1)) and k(cat) (0.33 ± 0.02 s(-1)), no sKIEs, ((D)(k(cat)/K(a)) = 1.05 ± 0.5 and (D)k(cat) = 1.06 ± 0.4), and pre-steady-state kinetics exhibited a discernible, but negligible, transient phase. For this third class of substrate, kinetic modeling was consistent with a mechanism in which k(dac) > k(ac) ∼ k(cat), and for which an isotope-insensitive step in the acylation half-reaction is the slowest. The combined results of these studies suggested that the identity of the amino acid at the P(1) position of the substrate is the main determinant of catalysis. On the basis of these kinetic data, together with crystallographic studies of substrate analogues and molecular dynamics analysis with models of acyl-enzyme intermediates, we present a catalytic model derived from the relative rates of the acylation vs deacylation half-reactions of cathepsin C. The chemical steps of catalysis are proposed to be dependent upon the conformational freedom of the amino acid substituents for optimal alignment for thiolation (acylation) or hydrolysis (deacylation). These studies suggest ideas for inhibitor design for papain-family cysteine proteases and strategies to progress drug discovery for other classes of disease-relevant cysteine proteases.


Subject(s)
Amino Acids/chemistry , Cathepsin C/chemistry , Dipeptides/chemistry , Catalysis , Cathepsin C/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Dynamics Simulation , Substrate Specificity
19.
Protein Expr Purif ; 76(1): 59-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20828618

ABSTRACT

Dipeptidyl peptidase I (DPPI) plays a crucial role in maturation of many regulatory peptides and has been suggested as a pharmaceutical target in several inflammatory diseases. It is also a useful processing enzyme for the generation of authentic protein products by catalyzing the removal of N-terminal fusion peptides. We used a robust transient transfection system in human embryonic kidney 293 cells to exploit expression and activation of DPPI from chicken, rat and man for the development of an industrial production process. The expression of human and rat DPPI was significantly higher in the human HEK293 cell line than that obtained with avian DPPI. A CHO K1SV stable cell line was selected as the optimal stable host system for production of human DPPI yielding expression levels higher than 1.5 g/L. The secreted pro-DPPI underwent auto-maturation during defined buffer conditions during the purification steps. Active human DPPI was purified with a three-step purification strategy employing: Butyl Sepharose 4 Fast Flow, Sephadex G-25 Medium and Q Sepharose Fast Flow chromatography. The final yield of active enzyme was approximately 1 g/L cell culture. The enzyme exhibited exopeptidase activity against both a dipeptide-p-nitroanilide substrate and N-terminally extended MEAE-hGH (Met-Glu-Ala-Glu-human growth hormone). In conclusion, an efficient production process for recombinant human DPPI has been developed including a highly efficient and stable CHO cell system and an efficient purification procedure, which is simple and easy to scale for industrial purposes. The present data facilitates not only industrial applications of DPPI as a processing enzyme, but also provides active enzyme useful in the identification of small molecule inhibitors.


Subject(s)
Cathepsin C/biosynthesis , Cathepsin C/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Animals , CHO Cells , Cathepsin C/chemistry , Chromogenic Compounds , Cricetinae , Cricetulus , Culture Media, Conditioned , Enzyme Activation , HEK293 Cells , Humans , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
20.
Chem Biol ; 17(8): 808-19, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797610

ABSTRACT

The widespread resistance of malaria parasites to all affordable drugs has made the identification of new targets urgent. Dipeptidyl aminopeptidases (DPAPs) represent potentially valuable new targets that are involved in hemoglobin degradation (DPAP1) and parasite egress (DPAP3). Here we use activity-based probes to demonstrate that specific inhibition of DPAP1 by a small molecule results in the formation of an immature trophozoite that leads to parasite death. Using computational methods, we designed stable, nonpeptidic covalent inhibitors that kill Plasmodium falciparum at low nanomolar concentrations. These compounds show signs of slowing parasite growth in a murine model of malaria, which suggests that DPAP1 might be a viable antimalarial target. Interestingly, we found that resynthesis and activation of DPAP1 after inhibition is rapid, suggesting that effective drugs would need to sustain DPAP1 inhibition for a period of 2-3 hr.


Subject(s)
Catalytic Domain , Cathepsin C/antagonists & inhibitors , Cathepsin C/metabolism , Computational Biology , Plasmodium falciparum/enzymology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Animals , Antiparasitic Agents/blood , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Cathepsin C/chemistry , Cell Line , Dose-Response Relationship, Drug , Drug Design , Drug Stability , Female , Malaria/drug therapy , Mice , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Protease Inhibitors/blood , Protease Inhibitors/therapeutic use , Trophozoites/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL