Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 268
1.
Br J Dermatol ; 188(1): 100-111, 2023 01 23.
Article En | MEDLINE | ID: mdl-36689511

BACKGROUND: Mendelian disorders of cornification (MeDOC) are a group of heterogeneous genodermatoses with different genetic bases. The pathogenesis of a substantial group of MeDOC remains to be elucidated. OBJECTIVES: To identify a new causative gene and the pathogenesis of a previously undescribed autosomal-dominant cornification disorder. METHODS: Whole-exome sequencing was performed in three families with the novel cornification disorder to identify the disease-causing variants. As the variants were located around the signal peptide (SP) cleavage site of a kallikrein-related peptidase, SP cleavage, subcellular localization and extracellular secretion of the variants were evaluated in eukaryotic overexpression systems by Western blotting or immunocytochemistry. Then the trypsin-like and chymotrypsin-like proteolytic activity of the peptidase and degradation of its catalytic substrate were assayed using the patients' stratum corneum (SC) samples. The morphology of the lamellar bodies and corneodesmosomes (CDs) in the patients' SC was ultrastructurally examined. A mouse model harbouring the equivalent variant was constructed and evaluated histologically. RESULTS: We identified two heterozygous variants affecting Gly50 in kallikrein-related peptidase (KLK)11 in a familial case and two sporadic cases with the new disorder, which is characterized by early-onset ichthyosiform erythroderma or erythrokeratoderma. KLK11 belongs to the family of kallikrein-related peptidases participating in skin desquamation by decomposing CDs, a process essential for shedding of the SC. In vitro experiments demonstrated that the variants perturbed the SP cleavage of KLK11, leading to subcellular mislocalization and impaired extracellular secretion of the KLK11 Gly50Glu variant. Both trypsin-like and chymotrypsin-like proteolytic activities were significantly decreased in the patients' SC samples. Reduced proteolysis of desmoglein 1 and delayed degeneration of CDs were detected in patients' SC, indicating delayed skin desquamation. Consistently, the patients showed a thickened, dense SC, indicating abnormal skin desquamation. Mice harbouring the homozygous c.131G>A (p.Gly44Glu) Klk11 variant, which is equivalent to KLK11 c.149G>A (p.Gly50Glu) in humans, exhibited hyperkeratosis and abnormal desquamation, partially recapitulating the phenotype. CONCLUSIONS: We provide evidence that variants at Gly50 affecting the SP cleavage of KLK11 cause a new autosomal-dominant cornification disorder with abnormal desquamation. Our findings highlight the essential role of KLKs in maintaining homeostasis of skin keratinization and desquamation.


Chymotrypsin , Protein Sorting Signals , Humans , Animals , Mice , Trypsin/metabolism , Chymotrypsin/metabolism , Kallikreins/chemistry , Kallikreins/metabolism , Skin/metabolism
2.
Cancer Biol Ther ; 23(1): 136-138, 2022 12 31.
Article En | MEDLINE | ID: mdl-35129066

The vast majority of the human proteome is yet to be functionally characterized thus hindering ongoing investigations on potential drug resistance mechanisms and advanced treatment options. Chemical proteomics is a powerful solution for enzyme profiling and the development of next generation cancer therapeutics previously deemed undruggable by small molecules. Within this field, activity-based protein profiling (ABPP) is a specialized technology capable of discriminating enzyme interactions that occur within complex, biological environments. In a recent publication by Lovell et al, the kallikrein-related peptidase (KLK) family of serine proteases that is highly implicated in the progression of prostate cancer (PCa) was subject to ABPP to elucidate enzymatic activities in the presence of enzalutamide. This is the first report of ABPP in PCa and of activity-based chemical probes selective for individual KLKs. Further, the study reveals androgen receptor-dependent activity among KLK proteins, particularly in mediating the invasion of the bone microenvironment.


Kallikreins , Prostatic Neoplasms , Humans , Kallikreins/chemistry , Kallikreins/metabolism , Kallikreins/therapeutic use , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Tumor Microenvironment
3.
Int J Mol Sci ; 22(23)2021 Dec 01.
Article En | MEDLINE | ID: mdl-34884832

The human kallikrein-related peptidase 4 (KLK4) and the transcribed pseudogene KLKP1 are reported to be highly expressed in the prostate. When trying to clone transcripts of KLKP1, we partly failed. Instead, we identified an androgen-regulated transcript, KLK4T2, which appeared to be a splice variant of KLK4 that also contained exons of KLKP1. Expression analysis of KLK4, KLK4T2, and KLKP1 transcripts in prostate cancer cell lines showed high levels of KLKP1 transcripts in the nucleus and in unfractionated cell extract, whereas it was almost completely absent in the cytoplasmatic fraction. This was in contrast to KLK4 and KLK4T2, which displayed high to moderate levels in the cytoplasm. In patient cohorts we found significantly higher expression of both KLK4T2 and KLK4 in benign prostatic hyperplasia compared to both primary prostate cancer and bone metastasis. Analysis of tissue panels demonstrated the highest expression of KLK4T2 in the prostate, but in contrast to the classical KLK4, relatively high levels were also found in placenta. So far, the function of KLK4T2 is still to be explored, but the structure of the translation product indicated that it generates a 17.4 kDa intracellular protein with possible regulatory function.


Kallikreins/metabolism , Alternative Splicing , Amino Acid Sequence , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Genetic Loci , Humans , Kallikreins/chemistry , Kallikreins/genetics , Male , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
J Biol Chem ; 297(5): 101342, 2021 11.
Article En | MEDLINE | ID: mdl-34710374

Peptide display methods are a powerful tool for discovering new ligands of pharmacologically relevant targets. However, the selected ligands often suffer from low affinity. Using phage display, we identified a new bicyclic peptide binder of prostate-specific membrane antigen (PSMA), a metalloprotease frequently overexpressed in prostate cancer. We show that linking multiple copies of a selected low-affinity peptide to a biocompatible water-soluble N-(2-hydroxypropyl)methacrylamide copolymer carrier (iBody) improved binding of the conjugate by several orders of magnitude. Furthermore, using ELISA, enzyme kinetics, confocal microscopy, and other approaches, we demonstrate that the resulting iBody can distinguish between different conformations of the target protein. The possibility to develop stable, fully synthetic, conformation-selective antibody mimetics has potential applications for molecular recognition, diagnosis and treatment of many pathologies. This strategy could significantly contribute to more effective drug discovery and design.


Biomimetic Materials/chemistry , Drug Carriers/chemistry , Peptide Library , Humans , Kallikreins/chemistry , Prostate-Specific Antigen/chemistry
5.
Acta Crystallogr D Struct Biol ; 77(Pt 8): 1084-1098, 2021 Aug 01.
Article En | MEDLINE | ID: mdl-34342281

Structures of BbKI, a recombinant Kunitz-type serine protease inhibitor from Bauhinia bauhinioides, complexed with human kallikrein 4 (KLK4) were determined at medium-to-high resolution in four crystal forms (space groups P3121, P6522, P21 and P61). Although the fold of the protein was virtually identical in all of the crystals, some significant differences were observed in the conformation of Arg64 of BbKI, the residue that occupies the S1 pocket in KLK4. Whereas this residue exhibited two orientations in the highest resolution structure (P3121), making either a canonical trypsin-like interaction with Asp189 of KLK4 or an alternate interaction, only a single alternate orientation was observed in the other three structures. A neighboring disulfide, Cys191-Cys220, was partially or fully broken in all KLK4 structures. Four variants of BbKI in which Arg64 was replaced by Met, Phe, Ala and Asp were expressed and crystallized, and their structures were determined in complex with KLK4. Structures of the Phe and Met variants complexed with bovine trypsin and of the Phe variant complexed with α-chymotrypsin were also determined. Although the inhibitory potency of these variant forms of BbKI was lowered by up to four orders of magnitude, only small changes were seen in the vicinity of the mutated residues. Therefore, a totality of subtle differences in KLK4-BbKI interactions within the fully extended interface in the structures of these variants might be responsible for the observed effect. Screening of the BbKI variants against a panel of serine proteases revealed an altered pattern of inhibitory specificity, which was shifted towards that of chymotrypsin-like proteases for the hydrophobic Phe and Met P1 substitutions. This work reports the first structures of plant Kunitz inhibitors with S1-family serine proteases other than trypsin, as well as new insights into the specificity of inhibition of medically relevant kallikreins.


Bauhinia/metabolism , Kallikreins/metabolism , Plant Proteins/metabolism , Kallikreins/chemistry , Mutation , Plant Proteins/chemistry , Protein Binding
6.
Sci Rep ; 11(1): 16170, 2021 08 09.
Article En | MEDLINE | ID: mdl-34373558

Proteinase-activated receptor-1 (PAR1), triggered by thrombin and other serine proteinases such as tissue kallikrein-4 (KLK4), is a key driver of inflammation, tumor invasiveness and tumor metastasis. The PAR1 transmembrane G-protein-coupled receptor therefore represents an attractive target for therapeutic inhibitors. We thus used a computational design to develop a new PAR1 antagonist, namely, a catalytically inactive human KLK4 that acts as a proteinase substrate-capture reagent, preventing receptor cleavage (and hence activation) by binding to and occluding the extracellular R41-S42 canonical PAR1 proteolytic activation site. On the basis of in silico site-saturation mutagenesis, we then generated KLK4S207A,L185D, a first-of-a-kind 'decoy' PAR1 inhibitor, by mutating the S207A and L185D residues in wild-type KLK4, which strongly binds to PAR1. KLK4S207A,L185D markedly inhibited PAR1 cleavage, and PAR1-mediated MAPK/ERK activation as well as the migration and invasiveness of melanoma cells. This 'substrate-capturing' KLK4 variant, engineered to bind to PAR1, illustrates proof of principle for the utility of a KLK4 'proteinase substrate capture' approach to regulate proteinase-mediated PAR1 signaling.


Kallikreins/metabolism , Receptor, PAR-1/antagonists & inhibitors , Amino Acid Substitution , Binding Sites , Cell Line, Tumor , Computer Simulation , Drug Design , Humans , Kallikreins/chemistry , Kallikreins/genetics , Kinetics , MCF-7 Cells , Mutagenesis, Site-Directed , Neoplasm Invasiveness/prevention & control , Protein Engineering , Protein Interaction Domains and Motifs , Proteolysis , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Substrate Specificity , Thrombin/metabolism
7.
J Med Chem ; 64(11): 7853-7876, 2021 06 10.
Article En | MEDLINE | ID: mdl-34044534

The contact system comprises a series of serine proteases that mediate procoagulant and proinflammatory activities via the intrinsic pathway of coagulation and the kallikrein-kinin system, respectively. Inhibition of Factor XIIa (FXIIa), an initiator of the contact system, has been demonstrated to lead to thrombo-protection and anti-inflammatory effects in animal models and serves as a potentially safer target for the development of antithrombotics. Herein, we describe the use of the Randomised Nonstandard Peptide Integrated Discovery (RaPID) mRNA display technology to identify a series of potent and selective cyclic peptide inhibitors of FXIIa. Cyclic peptides were evaluated in vitro, and three lead compounds exhibited significant prolongation of aPTT, a reduction in thrombin generation, and an inhibition of bradykinin formation. We also describe our efforts to identify the critical residues for binding FXIIa through alanine scanning, analogue generation, and via in silico methods to predict the binding mode of our lead cyclic peptide inhibitors.


Factor XIIa/antagonists & inhibitors , Peptides, Cyclic/chemistry , RNA, Messenger/metabolism , Serine Proteinase Inhibitors/chemistry , Binding Sites , Factor XIIa/metabolism , Gene Library , Genetic Code , Humans , Inhibitory Concentration 50 , Kallikreins/chemistry , Kallikreins/metabolism , Molecular Dynamics Simulation , Partial Thromboplastin Time , Peptides, Cyclic/metabolism , Protein Stability , Prothrombin Time , Puromycin/chemistry , RNA, Messenger/chemistry , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship
8.
Int J Mol Sci ; 22(5)2021 Mar 08.
Article En | MEDLINE | ID: mdl-33800517

A new PSMA ligand (PSMA-D4) containing the Glu-CO-Lys pharmacophore connected with a new linker system (L-Trp-4-Amc) and chelator DOTA was developed for radiolabeling with therapeutic radionuclides. Herein we describe the synthesis, radiolabeling, and preliminary biological evaluation of the novel PSMA-D4 ligand. Synthesized PSMA-D4 was characterized using TOF-ESI-MS, NMR, and HPLC methods. The novel compound was subject to molecular modeling with GCP-II to compare its binding mode to analogous reference compounds. The radiolabeling efficiency of PSMA-D4 with 177Lu, 90Y, 47Sc, and 225Ac was chromatographically tested. In vitro studies were carried out in PSMA-positive LNCaP tumor cells membranes. The ex vivo tissue distribution profile of the radioligands and Cerenkov luminescence imaging (CLI) was studied in LNCaP tumor-bearing mice. PSMA-D4 was synthesized in 24% yield and purity >97%. The radio complexes were obtained with high yields (>97%) and molar activity ranging from 0.11 to 17.2 GBq mcmol-1, depending on the radionuclide. In vitro assays confirmed high specific binding and affinity for all radiocomplexes. Biodistribution and imaging studies revealed high accumulation in LNCaP tumor xenografts and rapid clearance of radiocomplexes from blood and non-target tissues. These render PSMA-D4 a promising ligand for targeted therapy of prostate cancer (PCa) metastases.


Drug Delivery Systems , Kallikreins , Prostate-Specific Antigen , Prostatic Neoplasms , Radiopharmaceuticals , Animals , Humans , Kallikreins/chemistry , Kallikreins/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , Prostate-Specific Antigen/chemistry , Prostate-Specific Antigen/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article En | MEDLINE | ID: mdl-33397811

Kallikrein (PKa), generated by activation of its precursor prekallikrein (PK), plays a role in the contact activation phase of coagulation and functions in the kallikrein-kinin system to generate bradykinin. The general dogma has been that the contribution of PKa to the coagulation cascade is dependent on its action on FXII. Recently this dogma has been challenged by studies in human plasma showing thrombin generation due to PKa activity on FIX and also by murine studies showing formation of FIXa-antithrombin complexes in FXI deficient mice. In this study, we demonstrate high-affinity binding interactions between PK(a) and FIX(a) using surface plasmon resonance and show that these interactions are likely to occur under physiological conditions. Furthermore, we directly demonstrate dose- and time-dependent cleavage of FIX by PKa in a purified system by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis and chromogenic assays. By using normal pooled plasma and a range of coagulation factor-deficient plasmas, we show that this action of PKa on FIX not only results in thrombin generation, but also promotes fibrin formation in the absence of FXII or FXI. Comparison of the kinetics of either FXIa- or PKa-induced activation of FIX suggest that PKa could be a significant physiological activator of FIX. Our data indicate that the coagulation cascade needs to be redefined to indicate that PKa can directly activate FIX. The circumstances that drive PKa substrate specificity remain to be determined.


Bradykinin/metabolism , Factor IX/metabolism , Factor XII/metabolism , Fibrin/metabolism , Kallikreins/metabolism , Thrombin/metabolism , Blood Coagulation/physiology , Bradykinin/chemistry , Calcium/chemistry , Calcium/metabolism , Cations, Divalent , Factor IX/chemistry , Factor XI/chemistry , Factor XI/metabolism , Factor XII/chemistry , Fibrin/chemistry , Humans , Kallikreins/chemistry , Kinetics , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Protein Binding , Thrombin/chemistry
10.
Biomed Pharmacother ; 132: 110878, 2020 Dec.
Article En | MEDLINE | ID: mdl-33113419

Cancer is known to be one of the most major issues all around the world and is the most important cause of death. Prostate cancer is one of the most prevalent cancers among men, and the principal reason of death due to this cancer is the inappropriate detecting tools. Therefore, there is a great request for accurate diagnosis of prostate-specific antigen (PSA). Bio-analysis based on biomarkers might help to overcome this problem. Aptamers can be employed as high-affinity tools for cancer detection. The utilization of aptamer-based strategy in cancer investigation has demonstrated new horizons in biotechnology. The use of nanotechnology in biosensing is a serious development in this field. Advanced nanomaterials enhance the signal amplification in the biosensors, which also reduce the time required for diagnosis and analysis, they are also affordable, with high accuracy. In the present review (with 108 references), we discussed excellent features of the aptasensors on the sensitive and accurate monitoring of PSA biomarkers. Moreover, various types of nanomaterial-based aptasensors were surveyed for PSA detection (electrochemical, optical, piezoelectric, photoelectrochemical, electrochemiluminescent, and so forth). Furthermore, we reported the role of advanced nanomaterials, for instance graphene oxide, carbine nanotube, quantum dots, silica, gold, silver, and magnetic nanoparticles on the improvement of aptasensors of PSA. Finally, we discussed the advantages and limitations of different strategies on the early stage diagnosis of cancer. This article has been updated until July 2020.


Aptamers, Peptide/chemistry , Biosensing Techniques , Early Detection of Cancer , Kallikreins/blood , Nanomedicine , Nanoparticles , Prostate-Specific Antigen/blood , Prostatic Neoplasms/diagnosis , SELEX Aptamer Technique , Animals , Graphite/chemistry , Humans , Kallikreins/chemistry , Male , Metal Nanoparticles , Predictive Value of Tests , Prostate-Specific Antigen/chemistry , Prostatic Neoplasms/blood , Quantum Dots , Reproducibility of Results , Silicon Dioxide/chemistry
11.
Int J Mol Sci ; 21(12)2020 Jun 19.
Article En | MEDLINE | ID: mdl-32575583

Kallikrein-related peptidases (KLKs) and matrix metalloproteinases (MMPs) are secretory proteinases known to proteolytically process components of the extracellular matrix, modulating the pericellular environment in physiology and in pathologies. The interconnection between these families remains elusive. To assess the cross-activation of these families, we developed a peptide, fusion protein-based exposition system (Cleavage of exposed amino acid sequences, CleavEx) aiming at investigating the potential of KLK14 to recognize and hydrolyze proMMP sequences. Initial assessment identified ten MMP activation domain sequences which were validated by Edman degradation. The analysis revealed that membrane-type MMPs (MT-MMPs) are targeted by KLK14 for activation. Correspondingly, proMMP14-17 were investigated in vitro and found to be effectively processed by KLK14. Again, the expected neo-N-termini of the activated MT-MMPs was confirmed by Edman degradation. The effectiveness of proMMP activation was analyzed by gelatin zymography, confirming the release of fully active, mature MT-MMPs upon KLK14 treatment. Lastly, MMP14 was shown to be processed on the cell surface by KLK14 using murine fibroblasts overexpressing human MMP14. Herein, we propose KLK14-mediated selective activation of cell-membrane located MT-MMPs as an additional layer of their regulation. As both, KLKs and MT-MMPs, are implicated in cancer, their cross-activation may constitute an important factor in tumor progression and metastasis.


Enzyme Precursors/metabolism , Kallikreins/genetics , Kallikreins/metabolism , Matrix Metalloproteinase 14/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hydrolysis , Kallikreins/chemistry , Matrix Metalloproteinase 14/genetics , Mice , Porphyromonas gingivalis , Protein Engineering , Recombinant Proteins/metabolism
12.
Molecules ; 25(5)2020 Mar 02.
Article En | MEDLINE | ID: mdl-32131399

Despite promising anti-cancer properties in vitro, all titanium-based pharmaceuticals have failed in vivo. Likewise, no target-specific positron emission tomography (PET) tracer based on the radionuclide 45Ti has been developed, notwithstanding its excellent PET imaging properties. In this contribution, we present liquid-liquid extraction (LLE) in flow-based recovery and the purification of 45Ti, computer-aided design, and the synthesis of a salan-natTi/45Ti-chelidamic acid (CA)-prostate-specific membrane antigen (PSMA) ligand containing the Glu-urea-Lys pharmacophore. The compound showed compromised serum stability, however, no visible PET signal from the PC3+ tumor was seen, while the ex vivo biodistribution measured the tumor accumulation at 1.1% ID/g. The in vivo instability was rationalized in terms of competitive citrate binding followed by Fe(III) transchelation. The strategy to improve the in vivo stability by implementing a unimolecular ligand design is presented.


Computer Simulation , Neoplasms, Experimental/diagnostic imaging , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Titanium , Animals , Glutamic Acid/chemistry , Glutamic Acid/pharmacology , Humans , Kallikreins/chemistry , Kallikreins/pharmacokinetics , Kallikreins/pharmacology , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , PC-3 Cells , Prostate-Specific Antigen/chemistry , Prostate-Specific Antigen/pharmacokinetics , Prostate-Specific Antigen/pharmacology , Prostatic Neoplasms/metabolism , Pyridones/chemistry , Pyridones/pharmacokinetics , Pyridones/pharmacology , Radioactive Tracers , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Titanium/chemistry , Titanium/pharmacokinetics , Titanium/pharmacology , Urea/chemistry , Urea/pharmacology
13.
Thromb Haemost ; 120(3): 400-411, 2020 Mar.
Article En | MEDLINE | ID: mdl-31940673

Factor XII (FXII) zymogen activation requires cleavage after arginine 353 located in the activation loop. This cleavage can be executed by activated FXII (autoactivation), plasma kallikrein (PKa), or plasmin. Previous studies proposed that the activation loop of FXII is shielded to regulate FXII activation and subsequent contact activation. In this study, we aimed to elucidate this mechanism by expressing and characterizing seven consecutive N-terminally truncated FXII variants as well as full-length wild-type (WT) FXII. As soon as the fibronectin type II domain is lacking (FXII Δ1-71), FXII cleavage products appear on Western blot. These fragments display spontaneous amidolytic activity, indicating that FXII without the fibronectin type II domain is susceptible to autoactivation. Additionally, truncated FXII Δ1-71 is more easily activated by PKa or plasmin than full-length WT FXII. To exclude a contribution of autoactivation, we expressed active-site incapacitated FXII truncation variants (S544A). FXII S544A Δ1-71 is highly susceptible to cleavage by PKa, indicating exposure of the activation loop. In surface binding experiments, we found that the fibronectin type II domain is non-essential for binding to kaolin or polyphosphate, whereas the following epidermal growth factor-like domain is indispensable. Binding of full-length FXII S544A to kaolin or polyphosphate increases its susceptibility to cleavage by PKa. Moreover, the activation of full-length WT FXII by PKa increases approximately threefold in the presence of kaolin. Deletion of the fibronectin type II domain eliminates this effect. Combined, these findings suggest that the fibronectin type II domain shields the activation loop of FXII, ensuring zymogen quiescence.


Enzyme Precursors/chemistry , Factor XII/chemistry , Fibrinolysin/chemistry , Fibronectins/chemistry , Kallikreins/chemistry , Animals , Binding Sites , Blood Coagulation , Bradykinin/chemistry , Catalytic Domain , Cattle , Factor XIIa/chemistry , Fibronectins/blood , HEK293 Cells , Humans , Kallikreins/blood , Kaolin/chemistry , Polyphosphates/chemistry , Protein Binding , Protein Domains
14.
Allergy ; 75(2): 403-411, 2020 02.
Article En | MEDLINE | ID: mdl-31407378

BACKGROUND: Skin desquamation is facilitated by serine proteases KLK5 and KLK7, which are tightly regulated by lympho-epithelial Kazal-type related inhibitor (LEKTI). LEKTI itself is controlled through degraded by mesotrypsin. Here, we sought to determine whether LEKTI exonic mutations associated with atopic dermatitis (AD) affect the protease inhibitory activity of LEKTI or its susceptibility to mesotrypsin degradation. METHODS: The inhibitory activities of the LEKTI domain 4 (D4) and D6 WT and AD-associated mutants on the enzyme activities of KLK5 and KLK7 were compared using fluorogenic substrates. A keratinocyte cell culture system using HaCat cells was established to assess the role of D6 WT and D386N on triggering inflammation via the induction of thymic stromal lymphopoietin (TSLP). A degradation assay was used to assess the susceptibility of D4 and D6 mutants to mesotrypsin degradation. RESULTS: Enzymatic assays revealed that the D6 D386N mutation affected the inhibitory activity of LEKTI on KLK5 but not KLK7. Other exonic mutations on D6 (N368S, V395M, and E420K) and D4 (R267Q) did not alter LEKTI inhibition. The D386N mutation disrupted the role of D6 in suppressing TSLP induction by KLK5 in HaCat cells. Although WT D4 is more susceptible to mesotrypsin degradation than WT D6, the D4 R267Q mutant was more resistant to mesotrypsin degradation, whereas the D6 E420K mutant showed enhanced mesotrypsin-mediated degradation. CONCLUSION: Exonic mutations in D6, which previously have been associated with AD, may cause a disruption of inhibitory activity on KLK5 or enhance the degradation by mesotrypsin.


Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Exons , Mutation , Proteolysis , Serine Peptidase Inhibitor Kazal-Type 5/genetics , Cytokines/metabolism , Dermatitis, Atopic/pathology , Epidermis/metabolism , HaCaT Cells , Haplotypes , Humans , Inflammation/metabolism , Kallikreins/chemistry , Kallikreins/metabolism , Keratinocytes/metabolism , Polymorphism, Single Nucleotide , Protein Domains/genetics , Serine Peptidase Inhibitor Kazal-Type 5/metabolism , Signal Transduction/genetics , Trypsin/metabolism
15.
Neoplasia ; 21(10): 989-1002, 2019 10.
Article En | MEDLINE | ID: mdl-31446281

We present the functional characterization of a pseudogene associated recurrent gene fusion in prostate cancer. The fusion gene KLK4-KLKP1 is formed by the fusion of the protein coding gene KLK4 with the noncoding pseudogene KLKP1. Screening of a cohort of 659 patients (380 Caucasian American; 250 African American, and 29 patients from other races) revealed that the KLK4-KLKP1 is expressed in about 32% of prostate cancer patients. Correlative analysis with other ETS gene fusions and SPINK1 revealed a concomitant expression pattern of KLK4-KLKP1 with ERG and a mutually exclusive expression pattern with SPINK1, ETV1, ETV4, and ETV5. Development of an antibody specific to KLK4-KLKP1 fusion protein confirmed the expression of the full-length KLK4-KLKP1 protein in prostate tissues. The in vitro and in vivo functional assays to study the oncogenic properties of KLK4-KLKP1 confirmed its role in cell proliferation, cell invasion, intravasation, and tumor formation. Presence of strong ERG and AR binding sites located at the fusion junction in KLK4-KLKP1 suggests that the fusion gene is regulated by ERG and AR. Correlative analysis of clinical data showed an association of KLK4-KLKP1 with lower preoperative PSA values and in young men (<50 years) with prostate cancer. Screening of patient urine samples showed that KLK4-KLKP1 can be detected noninvasively in urine. Taken together, we present KLK4-KLKP1 as a class of pseudogene associated fusion transcript in cancer with potential applications as a biomarker for routine screening of prostate cancer.


Gene Fusion , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Pseudogenes , Amino Acid Sequence , Animals , Cell Line, Tumor , Chick Embryo , Gene Expression Regulation, Neoplastic , Genetic Loci , Humans , Kallikreins/chemistry , Kallikreins/genetics , Male , Neoplasm Grading , Oncogene Proteins, Fusion/chemistry , Tissue Kallikreins/chemistry , Tissue Kallikreins/genetics
16.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 8): 543-546, 2019 Aug 01.
Article En | MEDLINE | ID: mdl-31397325

Kallikrein 4 (KLK4) is a serine protease that is predominantly expressed in the prostate and is overexpressed in prostate cancer. As such, it has gained attention as an attractive target for prostate cancer therapeutics. Currently, only liganded structures of KLK4 exist in the Protein Data Bank. Until now, inferences about the subtle structural changes in KLK4 upon ligand binding have been made by comparison to other liganded forms, rather than to an apo form. In this study, an inhibitor-free form of KLK4 was crystallized. The crystals obtained belonged to space group P1, contained four molecules in the asymmetric unit and diffracted to 1.64 Šresolution. Interestingly, a nonstandard rotamer of the specificity-determining residue Asp189 was observed in all chains. This model will provide a useful unliganded structure for the future structure-guided design of KLK4 inhibitors.


Crystallography, X-Ray/methods , Kallikreins/chemistry , Kallikreins/metabolism , Humans , Models, Molecular , Protein Conformation , Substrate Specificity
17.
Pol J Vet Sci ; 22(2): 227-235, 2019 Jun.
Article En | MEDLINE | ID: mdl-31269332

The aim of this study was to identify the proteoforms of albumin and kallikrein in stallion seminal plasma (SP), and to determine their correlations with sperm motility parameters. The experimental material consisted of ejaculates from 8 stallions, which were collected during the breeding and non-breeding seasons (BS and NBS, respectively). SP proteins were identified by 2-D PAGE and mass spectrometry (MALDI TOT/TOF MS). Sperm motility parameters were analyzed using the CASA system. Protein expression (integrated optical density-IOD) of albumin proteoforms 1 (ALB 1) and 2 (ALB 2) and kallikrein proteoforms 1 (KAL 1) and 2 (KAL 2) was correlated (p⟨0.05) with sperm motility parameters (total motility and progressive motility) during the BS. No significant correlations were found between the expression of albumin or kallikrein and sperm motility parameters during the NBS. The presence of correlations between the expression of ALB 1, ALB 2, KAL 1, KAL 2 and selected sperm motility parameters could suggest that the analyzed components of the SP belong to the group of fertility-associated proteins (FAPs).


Albumins/chemistry , Horses , Kallikreins/chemistry , Semen/chemistry , Sperm Motility/physiology , Albumins/genetics , Albumins/metabolism , Animals , Kallikreins/genetics , Kallikreins/metabolism , Male , Protein Isoforms
18.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 5): 385-391, 2019 May 01.
Article En | MEDLINE | ID: mdl-31045568

The inhibition of kallikrein 5 (KLK5) has been identified as a potential strategy for treatment of the genetic skin disorder Netherton syndrome, in which loss-of-function mutations in the SPINK5 gene lead to down-regulation of the endogenous inhibitor LEKTI-1 and profound skin-barrier defects with severe allergic manifestations. To aid in the development of a medicine for this target, an X-ray crystallographic system was developed to facilitate fragment-guided chemistry and knowledge-based drug-discovery approaches. Here, the development of a surrogate crystallographic system in place of KLK5, which proved to be challenging to crystallize, is described. The biochemical robustness of the crystallographic surrogate and the suitability of the system for the study of small nonpeptidic fragments and lead-like molecules are demonstrated.


Benzamidines/chemistry , Kallikreins/chemistry , Protease Inhibitors/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Benzamidines/pharmacology , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Drug Discovery , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/genetics , Kallikreins/metabolism , Kinetics , Models, Molecular , Mutation , Netherton Syndrome/drug therapy , Netherton Syndrome/enzymology , Protease Inhibitors/pharmacology , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera , Static Electricity , Substrate Specificity
19.
Biochemistry ; 58(21): 2524-2533, 2019 05 28.
Article En | MEDLINE | ID: mdl-31058493

Sunflower trypsin inhibitor (SFTI-1) is a 14 amino acid serine protease inhibitor. The dual antiparallel ß-sheet arrangement of SFTI-1 is stabilized by an N-terminal-C-terminal backbone cyclization and a further disulfide bridge to form a final bicyclic structure. This constrained structure is further rigidified by an extensive network of internal hydrogen bonds. Thus, the structure of SFTI-1 in solution resembles the protease-bound structure, reducing the entropic penalty upon protease binding. When cleaved at the scissile bond, it is thought that the rigidifying features of SFTI-1 maintain its structure, allowing the scissile bond to be reformed. The lack of structural plasticity for SFTI-1 is proposed to favor initial protease binding and continued occupancy in the protease active site, resulting in an equilibrium between the cleaved and uncleaved inhibitor in the presence of a protease. We have determined, at 1.15 Å resolution, the X-ray crystal structures of complexes between human kallikrein-related peptidase 4 (KLK4) and SFTI-FCQR(Asn14) and between KLK4 and an acyclic form of the same inhibitor, SFTI-FCQR(Asn14)[1,14], with the latter displaying a cleaved scissile bond. Structural analysis and MD simulations together reveal the roles of the altered contact sequence, intramolecular hydrogen bonding network, and backbone cyclization in altering the state of SFTI's scissile bond ligation at the protease active site. Taken together, the data presented reveal insights into the role of dynamics in the standard-mechanism inhibition and suggest that modifications on the non-contact strand may be a useful, underexplored approach for generating further potent or selective SFTI-based inhibitors against members of the serine protease family.


Kallikreins/chemistry , Peptides, Cyclic/chemistry , Plant Proteins/chemistry , Serine Proteinase Inhibitors/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Cyclization , Escherichia coli/metabolism , Humans , Hydrogen Bonding , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Models, Molecular , Molecular Dynamics Simulation , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Plant Proteins/pharmacology , Protein Binding , Protein Conformation, beta-Strand , Serine Proteinase Inhibitors/pharmacology , Spodoptera/cytology , Spodoptera/metabolism , Transfection
20.
Comput Biol Chem ; 80: 433-440, 2019 Jun.
Article En | MEDLINE | ID: mdl-31146119

High production cost, instability, low tumor penetration are some of the shortcomings that have characterized and undermined the use of antibodies as a target for Cytotoxic T-lymphocytes associated protein 4 (CTLA-4). Design and discovery of small molecule inhibitors have therefore become a sine qua non in targeting immune proteins implicated in immune disorders. In this study, we utilized a drug repositioning approach to explore the characteristic feature of unrelated proteins to have similar binding sites and the promiscuity of drugs to repurpose an existing drug to target CTLA-4. CTLA-4 and Kallikrein-7 were found to have similar binding sites, we therefore used 1, 3, 6-trisubstituted 1, 4-diazepane-7-ones (TDSO) which is an inhibitor of Kallikrein-7 as our lead compound. High throughput screening using TDSO as a lead compound resulted in 9 hits with ZINC04515726 and ZINC08985213 having the highest binding score. We went ahead to investigate the interaction of these compounds with CTLA-4 by conducting a molecular dynamic simulation. Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) estimations revealed that TDSO had the highest binding energy value of -28.51Kcal/mol, with ZINC04515726 and ZINC08985213 having -23.76Kcal/mol and -21.03Kcal/mol respectively. The per-residue decomposition highlighted Tyr24, Ala25, Gly28, Ala30, Tyr53 and Asn72 as having significantly high electrostatic energy contributions and the main contributing residues to the binding of TDSO, ZINC04515726 and ZINC08985213 to Cytotoxic T lymphocytes CTLA-4. Summarily, from the results gathered, we proposed that TDSO can be an effective immune check point small molecule inhibitor against the suppression of T-cell activation, proliferation, and tumor cell eradication.


Azepines/metabolism , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , Drug Repositioning , Polypharmacology , Amino Acid Sequence , Azepines/chemistry , Azepines/pharmacokinetics , Binding Sites , CTLA-4 Antigen/chemistry , High-Throughput Screening Assays , Humans , Kallikreins/chemistry , Molecular Dynamics Simulation , Protein Binding
...