Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Nucl Med ; 65(7): 1051-1056, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38782459

ABSTRACT

Despite the inclusion of multiple agents within the prostate cancer treatment landscape, new treatment options are needed to address the unmet need for patients with metastatic castration-resistant prostate cancer (mCRPC). Although prostate-specific membrane antigen is the only cell-surface target to yield clinical benefit in men with advanced prostate cancer, additional targets may further advance targeted immune, cytotoxic, radiopharmaceutical, and other tumor-directed therapies for these patients. Human kallikrein 2 (hK2) is a novel prostate-specific target with little to no expression in nonprostate tissues. This first-in-human phase 0 trial uses an 111In-radiolabeled anti-hK2 monoclonal antibody, [111In]-DOTA-h11B6, to credential hK2 as a potential target for prostate cancer treatment. Methods: Participants with progressive mCRPC received a single infusion of 2 mg of [111In]-DOTA-h11B6 (185 MBq of 111In), with or without 8 mg of unlabeled h11B6 to assess antibody mass effects. Sequential imaging and serial blood samples were collected to determine [111In]-DOTA-h11B6 biodistribution, dosimetry, serum radioactivity, and pharmacokinetics. Safety was assessed within a 2-wk follow-up period from the time of [111In]-DOTA-h11B6 administration. Results: Twenty-two participants received [111In]-DOTA-h11B6 and are included in this analysis. Within 6-8 d of administration, [111In]-DOTA-h11B6 visibly accumulated in known mCRPC lesions, with limited uptake in other organs. Two treatment-emergent adverse events unrelated to treatment occurred, including tumor-related bleeding in 1 patient, which led to early study discontinuation. Serum clearance, biodistribution, and tumor targeting were independent of total antibody mass (2 or 10 mg). Conclusion: This first-in-human study demonstrates that tumor-associated hK2 can be identified and targeted using h11B6 as a platform as the h11B6 antibody selectively accumulated in mCRPC metastases with mass-independent clearance kinetics. These data support the feasibility of hK2 as a target for imaging and hK2-directed agents as potential therapies in patients with mCRPC.


Subject(s)
Neoplasm Metastasis , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Tissue Distribution , Aged , Middle Aged , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Tissue Kallikreins/antagonists & inhibitors , Indium Radioisotopes , Isotope Labeling , Heterocyclic Compounds, 1-Ring/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use
2.
J Am Heart Assoc ; 10(5): e019372, 2021 02.
Article in English | MEDLINE | ID: mdl-33599139

ABSTRACT

Background Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. The kinin B2 receptor agonist, bradykinin, has been implicated in AAA pathogenesis through promoting inflammation. Bradykinin is generated from high- and low-molecular-weight kininogen by the serine protease kallikrein-1. The aims of this study were first to examine the effect of neutralizing kallikrein-1 on AAA development in a mouse model and second to test how blocking kallikrein-1 affected cyclooxygenase-2 and prostaglandin E2 in human AAA explants. Methods and Results Neutralization of kallikrein-1 in apolipoprotein E-deficient (ApoE-/-) mice via administration of a blocking antibody inhibited suprarenal aorta expansion in response to angiotensin (Ang) II infusion. Kallikrein-1 neutralization decreased suprarenal aorta concentrations of bradykinin and prostaglandin E2 and reduced cyclooxygenase-2 activity. Kallikrein-1 neutralization also decreased protein kinase B and extracellular signal-regulated kinase 1/2 phosphorylation and reduced levels of active matrix metalloproteinase 2 and matrix metalloproteinase 9. Kallikrein-1 blocking antibody reduced levels of cyclooxygenase-2 and secretion of prostaglandin E2 and active matrix metalloproteinase 2 and matrix metalloproteinase 9 from human AAA explants and vascular smooth muscle cells exposed to activated neutrophils. Conclusions These findings suggest that kallikrein-1 neutralization could be a treatment target for AAA.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/therapy , Dinoprostone/metabolism , Muscle, Smooth, Vascular/pathology , Tissue Kallikreins/antagonists & inhibitors , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Biopsy , Cells, Cultured , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism
3.
Biomolecules ; 10(6)2020 05 28.
Article in English | MEDLINE | ID: mdl-32481593

ABSTRACT

Kallistatin, also known as SERPINA4, has been implicated in the regulation of blood pressure and angiogenesis, due to its specific inhibition of tissue kallikrein 1 (KLK1) and/or by its heparin binding ability. The binding of heparin on kallistatin has been shown to block the inhibition of KLK1 by kallistatin but the detailed molecular mechanism underlying this blockade is unclear. Here we solved the crystal structures of human kallistatin and its complex with heparin at 1.9 and 1.8 Å resolution, respectively. The structures show that kallistatin has a conserved serpin fold and undergoes typical stressed-to-relaxed conformational changes upon reactive loop cleavage. Structural analysis and mutagenesis studies show that the heparin binding site of kallistatin is located on a surface with positive electrostatic potential near a unique protruded 310 helix between helix H and strand 2 of ß-sheet C. Heparin binding on this site would prevent KLK1 from docking onto kallistatin due to the electrostatic repulsion between heparin and the negatively charged surface of KLK1, thus blocking the inhibition of KLK1 by kallistatin. Replacement of the acidic exosite 1 residues of KLK1 with basic amino acids as in thrombin resulted in accelerated inhibition. Taken together, these data indicate that heparin controls the specificity of kallistatin, such that kinin generation by KLK1 within the microcirculation will be locally protected by the binding of kallistatin to the heparin-like glycosaminoglycans of the endothelium.


Subject(s)
Heparin/pharmacology , Serpins/metabolism , Static Electricity , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/metabolism , Humans
4.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1127-L1140, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30908937

ABSTRACT

Host cell proteases are involved in influenza pathogenesis. We examined the role of tissue kallikrein 1 (KLK1) by comparing wild-type (WT) and KLK1-deficient mice infected with influenza H3N2 virus. The levels of KLK1 in lung tissue and in bronchoalveolar lavage (BAL) fluid increased substantially during infection. KLK1 did not promote virus infectivity despite its trypsin-like activity, but it did decrease the initial virus load. We examined two cell types involved in the early control of pathogen infections, alveolar macrophages (AMs) and natural killer (NK) cells to learn more about the antiviral action of KLK1. Inactivating the Klk1 gene or treating WT mice with an anti-KLK1 monoclonal antibody to remove KLK1 activity accelerated the initial virus-induced apoptotic depletion of AMs. Intranasal instillation of deficient mice with recombinant KLK1 (rKLK1) reversed the phenotype. The levels of granulocyte-macrophage colony-stimulating factor in infected BAL fluid were significantly lower in KLK1-deficient mice than in WT mice. Treating lung epithelial cells with rKLK1 increased secretion of this factor known to enhance AM resistance to pathogen-induced apoptosis. The recruitment of NK cells to the air spaces peaked 3 days after infection in WT mice but not in KLK1-deficient mice, as did increases in several NK-attracting chemokines (CCL2, CCL3, CCL5, and CXCL10) in BAL. Chronic obstructive pulmonary disease (COPD) patients are highly susceptible to viral infection, and we observed that the KLK1 mRNA levels decreased with increasing COPD severity. Our findings indicate that KLK1 intervenes early in the antiviral defense modulating the severity of influenza infection. Decreased KLK1 expression in COPD patients could contribute to the worsening of influenza.


Subject(s)
Apoptosis/physiology , Macrophages, Alveolar/pathology , Orthomyxoviridae Infections/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Tissue Kallikreins/metabolism , A549 Cells , Acute Lung Injury/pathology , Acute Lung Injury/virology , Animals , Cell Line , Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , Dogs , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , Influenza A Virus, H3N2 Subtype , Killer Cells, Natural/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Pulmonary Disease, Chronic Obstructive/virology , Respiratory Mucosa/metabolism , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/genetics
5.
PLoS One ; 11(11): e0166268, 2016.
Article in English | MEDLINE | ID: mdl-27824929

ABSTRACT

Tissue kallikreins (KLKs), in particular KLK5, 7 and 14 are the major serine proteases in the skin responsible for skin shedding and activation of inflammatory cell signaling. In the normal skin, their activities are controlled by an endogenous protein protease inhibitor encoded by the SPINK5 gene. Loss-of-function mutations in SPINK5 leads to enhanced skin kallikrein activities and cause the skin disease Netherton Syndrome (NS). We have been developing inhibitors based on the Sunflower Trypsin Inhibitor 1 (SFTI-1) scaffold, a 14 amino acids head-to-tail bicyclic peptide with a disulfide bond. To optimize a previously reported SFTI-1 analogue (I10H), we made five analogues with additional substitutions, two of which showed improved inhibition. We then combined those substitutions and discovered a variant (Analogue 6) that displayed dual inhibition of KLK5 (tryptic) and KLK7 (chymotryptic). Analogue 6 attained a tenfold increase in KLK5 inhibition potency with an Isothermal Titration Calorimetry (ITC) Kd of 20nM. Furthermore, it selectively inhibits KLK5 and KLK14 over seven other serine proteases. Its biological function was ascertained by full suppression of KLK5-induced Protease-Activated Receptor 2 (PAR-2) dependent intracellular calcium mobilization and postponement of Interleukin-8 (IL-8) secretion in cell model. Moreover, Analogue 6 permeates through the cornified layer of in vitro organotypic skin equivalent culture and inhibits protease activities therein, providing a potential drug lead for the treatment of NS.


Subject(s)
Helianthus/metabolism , Peptides, Cyclic/antagonists & inhibitors , Skin Diseases/drug therapy , Tissue Kallikreins/antagonists & inhibitors , Cell Line , Humans , Interleukin-8/metabolism , Netherton Syndrome/drug therapy , Netherton Syndrome/metabolism , Peptides, Cyclic/metabolism , Proteinase Inhibitory Proteins, Secretory/pharmacology , Receptor, PAR-2/metabolism , Skin/drug effects , Skin/metabolism , Skin Diseases/metabolism , Trypsin Inhibitors/pharmacology
6.
Expert Opin Ther Targets ; 20(7): 801-18, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26941073

ABSTRACT

INTRODUCTION: Tissue kallikrein and the kallikrein-related peptidases (KLKs) constitute a family of 15 homologous secreted serine proteases with trypsin- or chymotrypsin-like activities, which participate in a broad spectrum of physiological procedures. Deregulated expression and/or activation of the majority of the family members have been reported in several human diseases, thereby making KLKs ideal targets for therapeutic intervention. AREAS COVERED: In the present review, we summarize the role of KLKs in normal human physiology and pathology, focusing on prostate cancer and skin diseases. Furthermore, we discuss the recent advances in the development of KLK-based therapies. A great number of diverse engineered KLKs inhibitors with improved potency, selectivity and immunogenicity have been synthesized by redesigning examples that are endogenous and naturally occurring. Moreover, encouraging results have been documented using KLKs-based vaccines and immunotherapies, as well as KLKs-mediated activation of pro-drugs. Finally, KLKs-targeting aptamers and KLKs-based imaging tools represent novel approaches towards the exploitation of KLKs' therapeutic value. EXPERT OPINION: The central/critical roles of KLK family in several human pathologies highlight KLKs as attractive molecular targets for developing novel therapeutics.


Subject(s)
Kallikreins/antagonists & inhibitors , Prostatic Neoplasms/therapy , Skin Diseases/therapy , Animals , Drug Design , Humans , Immunotherapy/methods , Kallikreins/metabolism , Male , Molecular Targeted Therapy , Prodrugs , Prostatic Neoplasms/pathology , Skin Diseases/pathology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/metabolism , Vaccines/administration & dosage
7.
Bioorg Med Chem Lett ; 26(5): 1485-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26848109

ABSTRACT

The human tissue kallikreins (KLK1-KLK15) comprise a family of 15 serine peptidases detected in almost every tissue of the human body and that actively participate in many physiological and pathological events. Some kallikreins are involved in diseases for which no effective therapy is available, as for example, epithelial disorders, bacterial infections and in certain cancers metastatic processes. In recent years our group have made efforts to find inhibitors for all kallikreins, based on natural products and synthetic molecules, and all the inhibitors developed by our group presented a competitive mechanism of inhibition. Here we describe fukugetin, a natural product that presents a mixed-type mechanism of inhibition against KLK1 and KLK2. This type of inhibitor is gaining importance today, especially for the development of exosite-type inhibitors, which present potential to selectively inhibit the enzyme activity only against specific substrate.


Subject(s)
Biflavonoids/pharmacology , Biological Products/pharmacology , Serine Proteinase Inhibitors/pharmacology , Tissue Kallikreins/antagonists & inhibitors , Biflavonoids/chemistry , Biflavonoids/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Dose-Response Relationship, Drug , Garcinia/chemistry , Humans , Models, Molecular , Molecular Conformation , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/isolation & purification , Structure-Activity Relationship , Tissue Kallikreins/metabolism
8.
Nat Rev Drug Discov ; 14(3): 183-202, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25698643

ABSTRACT

Tissue kallikreins are a family of fifteen secreted serine proteases encoded by the largest protease gene cluster in the human genome. In the past decade, substantial progress has been made in characterizing the natural substrates, endogenous inhibitors and in vivo functions of kallikreins, and studies have delineated important pathophysiological roles for these proteases in a variety of tissues. Thus, kallikreins are now considered attractive targets for the development of novel therapeutics for airway, cardiovascular, tooth, brain, skin and neoplastic diseases. In this Review, we discuss recent advances in our understanding of the physiological functions and pathological implications of kallikrein proteases, and highlight progress in the identification of kallikrein inhibitors, which together are bringing us closer to therapeutically targeting kallikreins in selected disease settings.


Subject(s)
Serine Proteinase Inhibitors/therapeutic use , Tissue Kallikreins/antagonists & inhibitors , Animals , Humans , Models, Molecular , Serine Proteases/genetics , Serine Proteases/physiology , Tissue Kallikreins/genetics , Tissue Kallikreins/physiology
9.
Proteins ; 83(7): 1209-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25143259

ABSTRACT

Off-target binding connotes the binding of a small molecule of therapeutic significance to a protein target in addition to the primary target for which it was proposed. Progressively such off-targeting is emerging to be regular practice to reveal side effects. Chymase is an enzyme of hydrolase class that catalyzes hydrolysis of peptide bonds. A link between heart failure and chymase is ascribed, and a chymase inhibitor is in clinical phase II for treatment of heart failure. However, the underlying mechanisms of the off-target effects of human chymase inhibitors are still unclear. Here, we develop a robust computational strategy that is applicable to any enzyme system and that allows the prediction of drug effects on biological processes. Putative off-targets for chymase inhibitors were identified through various structural and functional similarity analyses along with molecular docking studies. Finally, literature survey was performed to incorporate these off-targets into biological pathways and to establish links between pathways and particular adverse effects. Off-targets of chymase inhibitors are linked to various biological pathways such as classical and lectin pathways of complement system, intrinsic and extrinsic pathways of coagulation cascade, and fibrinolytic system. Tissue kallikreins, granzyme M, neutrophil elastase, and mesotrypsin are also identified as off-targets. These off-targets and their associated pathways are elucidated for the effects of inflammation, cancer, hemorrhage, thrombosis, and central nervous system diseases (Alzheimer's disease). Prospectively, our approach is helpful not only to better understand the mechanisms of chymase inhibitors but also for drug repurposing exercises to find novel uses for these inhibitors.


Subject(s)
Chymases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Systems Biology/methods , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Blood Coagulation/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/pathology , Chymases/chemistry , Chymases/metabolism , Complement Pathway, Mannose-Binding Lectin/drug effects , Drug Design , Enzyme Inhibitors/pharmacology , Fibrinolysis/drug effects , Granzymes/antagonists & inhibitors , Granzymes/chemistry , Granzymes/metabolism , Humans , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/chemistry , Leukocyte Elastase/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/chemistry , Tissue Kallikreins/metabolism , Trypsin/chemistry , Trypsin/metabolism , User-Computer Interface
10.
Biochem Biophys Res Commun ; 449(1): 69-73, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24814709

ABSTRACT

Rhipicephalus microplus is an important ectoparasite that is responsible for transmission of anaplasmosis and babesiosis to cattle. Tissue kallikrein inhibitors might play an important role in R. microplus eggs. In the present work, we purified and characterized, a tissue kallikrein inhibitor presents in R. microplus eggs (RmKK), a protein which contains two Kunitz domain in tandem. Purified inhibitor was confirmed by amino terminal determination and its dissociation constant (Ki) for bovine trypsin and porcine pancreatic kallikrein were 0.6 nM and 91.5 nM, respectively. Using a cDNA library from R. microplus midgut, we cloned the cDNA fragment encoding mature RmKK and expressed the protein in Pichia pastoris system. Recombinant RmKK was purified by ion exchange chromatography and presented molecular mass of 16.3 kDa by MALDI-TOF analysis. Moreover, RmKK showed a tight binding inhibition for serine proteases as bovine trypsin (Ki=0.2 nM) and porcine pancreatic kallikrein (PPK) (Ki=300 nM). We performed, for the first time, the characterization of a tissue kallikrein inhibitor presents in R. microplus eggs, which the transcript is produced in the adult female gut. BmKK seems to be the strongest PPK inhibitor among all BmTIs present in the eggs and larvae (Andreotti et al., 2001; Sasaki et al., 2004). This data suggests that BmKK may participate in the development of tick egg and larvae phase.


Subject(s)
Arthropod Proteins/chemistry , Arthropod Proteins/metabolism , Ovum/metabolism , Rhipicephalus/classification , Rhipicephalus/metabolism , Tissue Kallikreins/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , Enzyme Activation , Female , Molecular Sequence Data , Protein Binding , Species Specificity , Tissue Distribution
11.
Expert Opin Ther Targets ; 18(4): 365-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24571737

ABSTRACT

INTRODUCTION: Novel therapeutic compounds are needed for prostate cancer (CaP), given the limitations of already used drugs and the disease's mortality, often attributed to castrate resistance. Tissue kallikrein and kallikrein-related peptidases (KLKs) form a family of serine proteases aberrantly expressed and broadly implicated in human malignancies. In CaP, KLKs participate in the promotion of cell proliferation, extracellular matrix degradation, tumour cell invasion and metastasis. AREAS COVERED: This review discusses the different ways of inhibiting, modulating and exploiting KLK activity and/or expression as emerging CaP therapeutics. KLKs are targeted by diverse naturally occurring substances, including proteinaceous inhibitors, low-molecular-weight peptides and Zn(2+). Synthetic KLK inhibitors include protein/peptide-based inhibitors and small molecules. A re-engineered serpin-based KLK inhibitor is under evaluation in first-in-human trials as a CaP therapeutic, whereas additional potent and selective KLK inhibitors with relevance to CaP have been synthesized. KLK3-activated pro-drugs have entered Phase I and Phase II clinical trials as therapeutics for prostate tumours. The KLK3-based PROSTVAC® vaccine is evaluated in Phase III clinical trials. Targeting KLK expression via RNA interference methods could represent another promising therapeutic approach for CaP. EXPERT OPINION: Apart from their immense biomarker potential, KLKs also hold promise as the basis of novel CaP therapeutics.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/trends , Prostatic Neoplasms/drug therapy , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/biosynthesis , Animals , Clinical Trials as Topic/methods , Drug Delivery Systems/methods , Humans , Male , Peptide Hydrolases/metabolism , Prostatic Neoplasms/enzymology , RNA Interference/physiology
12.
J Pharmacol Exp Ther ; 335(3): 681-92, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20847038

ABSTRACT

Inhaled bradykinin causes bronchoconstriction in asthmatic subjects but not nonasthmatics. To date, animal studies with inhaled bradykinin have been performed only in anesthetized guinea pigs and rats, where it causes bronchoconstriction through sensory nerve pathways. In the present study, airway function was recorded in conscious guinea pigs by whole-body plethysmography. Inhaled bradykinin (1 mM, 20 s) caused bronchoconstriction and influx of inflammatory cells to the lungs, but only when the enzymatic breakdown of bradykinin by angiotensin-converting enzyme and neutral endopeptidase was inhibited by captopril (1 mg/kg i.p.) and phosphoramidon (10 mM, 20-min inhalation), respectively. The bronchoconstriction and cell influx were antagonized by the B(2) kinin receptor antagonist 4-(S)-amino-5-(4-{4-[2,4-dichloro-3-(2,4-dimethyl-8-quinolyloxymethyl)phenylsulfonamido]-tetrahydro-2H-4-pyranylcarbonyl}piperazino)-5-oxopentyl](trimethyl)ammonium chloride hydrochloride (MEN16132) when given by inhalation (1 and 10 µM, 20 min) and are therefore mediated via B(2) kinin receptors. However, neither intraperitioneal MEN16132 nor the peptide B(2) antagonist icatibant, by inhalation, antagonized these bradykinin responses. Sensitization of guinea pigs with ovalbumin was not sufficient to induce airway hyperreactivity (AHR) to the bronchoconstriction by inhaled bradykinin. However, ovalbumin challenge of sensitized guinea pigs caused AHR to bradykinin and histamine. Infection of guinea pigs by nasal instillation of parainfluenza-3 virus produced AHR to inhaled histamine and lung influx of inflammatory cells. These responses were attenuated by the bradykinin B(2) receptor antagonist MEN16132 and H-(4-chloro)DPhe-2'(1-naphthylalanine)-(3-aminopropyl)guanidine (VA999024), an inhibitor of tissue kallikrein, the enzyme responsible for lung synthesis of bradykinin. These results suggest that bradykinin is involved in virus-induced inflammatory cell influx and AHR.


Subject(s)
Bradykinin/pharmacology , Bronchial Hyperreactivity/physiopathology , Bronchoconstriction/drug effects , Parainfluenza Virus 3, Human , Pneumonia/chemically induced , Respirovirus Infections/pathology , Respirovirus Infections/physiopathology , Animals , Bradykinin/administration & dosage , Bradykinin/antagonists & inhibitors , Bradykinin B2 Receptor Antagonists , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/cytology , Captopril/pharmacology , Cell Count , Glycopeptides/pharmacology , Guinea Pigs , Histamine/pharmacology , Male , Ornithine/analogs & derivatives , Ornithine/pharmacology , Ornithine/therapeutic use , Ovalbumin/immunology , Peptides/pharmacology , Peptides/therapeutic use , Plethysmography, Whole Body , Pneumonia/pathology , Protease Inhibitors/pharmacology , Respirovirus Infections/drug therapy , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tissue Kallikreins/antagonists & inhibitors
13.
Biol Chem ; 391(7): 803-12, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20482314

ABSTRACT

We recently demonstrated that tissue kallikrein (TK) promotes keratinocyte migration through activation of protease-activated receptor-1 (PAR(1)) and transactivation of the epi-dermal growth factor receptor (EGFR). In this study, we investigated the potential role of PAR(1) in mediating the effect of TK on cancer cell migration, invasion and proliferation. Our results show that TK promotes DU145 prostate cancer cell migration in a concentration-dependent manner, but has no effect on A549 lung cancer cells. Active TK markedly increases DU145 cell migration and invasion, which are blocked by aprotinin but minimally affected by icatibant; kinin treatment has little effect. TK-induced cell migration and invasion are abolished by inhibition of PAR(1) using a pharmacological inhibitor or RNA interference. The effect of TK on cell migration and invasion are also blocked by inhibitors of protein kinase C, c-Src, matrix metalloproteinase, EGFR and extracellular signal-regulated kinase (ERK). Moreover, TK stimulates ERK phosphorylation, which is inhibited by an EGFR antagonist. Additionally, TK but not kinin stimulates DU145 cell proliferation through activation of the kinin B2 receptor, but not PAR(1) and EGFR. These results indicate differential signaling pathways mediated by TK in promoting prostate cancer cell migration and invasion via PAR(1) activation, and proliferation via kinin B2 receptor stimulation.


Subject(s)
Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, PAR-1/metabolism , Tissue Kallikreins/metabolism , Aprotinin/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Kinins/pharmacology , Male , Neoplasm Invasiveness , Receptor, PAR-1/antagonists & inhibitors , Signal Transduction/drug effects , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/isolation & purification , Tumor Cells, Cultured
14.
Biol Chem ; 391(4): 475-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20180640

ABSTRACT

Kallikrein-related peptidase 2 (KLK2) degrades insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3) in vitro. IGFBP-3 forms complexes with IGFs, preventing them from binding to their receptors and stimulating cell proliferation and survival. IGF-independent actions have also been described for IGFBP-3. The degradation of IGFBP-3 by KLK2 or other proteases in the prostate may promote the growth of prostate cancer. We studied IGFBP-3 degradation by immunoblotting and two specific immunoassays, one recognizing only native non-fragmented IGFBP-3 and the other one recognizing both intact and proteolytically cleaved IGFBP-3. Peptides were used to inhibit the enzyme activity of KLK2 and cleavage sites in IGFBP-3 were identified by mass spectrometry. KLK2 proteolyzed IGFBP-3 into several small fragments, mostly after Arg residues, in keeping with the trypsin-like activity of KLK2. The fragmentation could be inhibited by KLK2-inhibiting peptides in a dose-dependent fashion. As degradation of IGFBP-3 could lead to a more aggressive cancer phenotype, inhibition of KLK2 activity might be useful for treatment of prostate cancer and other diseases associated with increased KLK2 activity.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/pharmacology , Serine Proteinase Inhibitors/pharmacology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/metabolism , Amino Acid Sequence , Animals , Fluoroimmunoassay , Humans , Immunoblotting , Mass Spectrometry , Molecular Sequence Data
15.
Biol Chem ; 391(4): 345-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20180644

ABSTRACT

Tissue kallikrein (KLK1) processes low-molecular weight kininogen to produce vasoactive kinins, which exert biological functions via kinin receptor signaling. Using various delivery approaches, we have demonstrated that tissue kallikrein through kinin B2 receptor signaling exhibits a wide spectrum of beneficial effects by reducing cardiac and renal injuries, restenosis and ischemic stroke, and by promoting angiogenesis and skin wound healing, independent of blood pressure reduction. Protection by tissue kallikrein in oxidative organ damage is attributed to the inhibition of apoptosis, inflammation, hypertrophy and fibrosis. Tissue kallikrein also enhances neovascularization in ischemic heart and limb. Moreover, tissue kallikrein/kinin infusion not only prevents but also reverses kidney injury, inflammation and fibrosis in salt-induced hypertensive rats. Furthermore, there is a wide time window for kallikrein administration in protection against ischemic brain infarction, as delayed kallikrein infusion for 24 h after cerebral ischemia in rats is effective in reducing neurological deficits, infarct size, apoptosis and inflammation. Importantly, in the clinical setting, human tissue kallikrein has been proven to be effective in the treatment of patients with acute brain infarction when injected within 48 h after stroke onset. Finally, kallikrein promotes skin wound healing and keratinocyte migration by direct activation of protease-activated receptor 1.


Subject(s)
Cardiovascular Diseases/metabolism , Cerebrovascular Disorders/metabolism , Kidney Diseases/metabolism , Skin/injuries , Skin/metabolism , Tissue Kallikreins/metabolism , Wound Healing , Animals , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/prevention & control , Cerebrovascular Disorders/physiopathology , Cerebrovascular Disorders/prevention & control , Humans , Kidney Diseases/prevention & control , Skin/pathology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/chemistry , Tissue Kallikreins/therapeutic use
16.
Am J Physiol Renal Physiol ; 298(4): F1033-40, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20089675

ABSTRACT

Levels of tissue kallikrein (TK) are significantly lower in the urine of patients with kidney failure, and TK expression is specifically diminished in rat kidney after recovery from ischemia-reperfusion injury. In this study, we investigated the functional consequence of blocking endogenous TK activity in a rat model of chronic kidney disease. Inhibition of endogenous TK levels for 10 days by neutralizing TK antibody injection in DOCA-salt rats caused a significant increase in blood urea nitrogen and urinary protein levels, and a decrease in creatinine clearance. Kidney sections from anti-TK antibody-treated rats displayed a marked rise in tubular dilation and protein cast accumulation as well as glomerular sclerosis and size. TK blockade also increased inflammatory cell infiltration, myofibroblast and collagen accumulation, and collagen fraction volume. Elevated renal inflammation and fibrosis by anti-TK antibody were associated with increased expression of tumor necrosis factor-alpha, intercellular adhesion molecule-1, tissue inhibitor of metalloproteinase-2 (TIMP-2), and plasminogen activator inhibitor-1 (PAI-1). Moreover, the detrimental effect of TK blockade resulted in reduced nitric oxide (NO) levels as well as increased serum lipid peroxidation, renal NADH oxidase activity, and superoxide formation. In cultured proximal tubular cells, TK inhibited angiotensin II-induced superoxide production and NADH oxidase activity via NO formation. In addition, TK markedly increased matrix metalloproteinase-2 activity with a parallel reduction of TIMP-2 and PAI-1 synthesis. These findings indicate that endogenous TK has the propensity to preserve kidney structure and function in rats with chronic renal disease by inhibiting oxidative stress and activating matrix degradation pathways.


Subject(s)
Kidney Diseases/metabolism , Tissue Kallikreins/antagonists & inhibitors , Angiotensin II , Animals , Antibodies , Cell Line , Humans , Kidney Tubules, Proximal/cytology , Male , Metalloproteases/metabolism , Oxidative Stress , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Rats , Rats, Inbred WF , Reactive Oxygen Species , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Kallikreins/metabolism , Transforming Growth Factor beta/pharmacology
17.
Biochem J ; 422(2): 383-92, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19527222

ABSTRACT

KLK1 (tissue kallikrein 1) is a member of the tissue kallikrein family of serine proteases and is the primary kinin-generating enzyme in human airways. DX-2300 is a fully human antibody that inhibits KLK1 via a competitive inhibition mechanism (Ki=0.13 nM). No binding of DX-2300 to KLK1 was observed in a surface-plasmon-resonance biosensor assay when KLK1 was complexed to known active-site inhibitors, suggesting that DX-2300 recognizes the KLK1 active site. DX-2300 did not inhibit any of the 21 serine proteases that were each tested at a concentration of 1 microM. We validated the use of DX-2300 for specific KLK1 inhibition by measuring the inhibition of KLK1-like activity in human urine, saliva and bronchoalveolar lavage fluid, which are known to contain active KLK1. In human tracheobronchial epithelial cells grown at the air/liquid interface, DX-2300 blocked oxidative-stress-induced epidermal-growth-factor receptor activation and downstream mucus cell proliferation and hypersecretion, which have been previously shown to be mediated by KLK1. In an allergic sheep model of asthma, DX-2300 inhibited both allergen-induced late-phase bronchoconstriction and airway hyper-responsiveness to carbachol. These studies demonstrate that DX-2300 is a potent and specific inhibitor of KLK1 that is efficacious in in vitro and in vivo models of airway disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , Asthma/enzymology , Asthma/therapy , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/therapy , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/physiology , Animals , Antibodies, Monoclonal/metabolism , Asthma/immunology , Binding Sites, Antibody , Binding, Competitive , Cells, Cultured , Humans , Mice , Pulmonary Disease, Chronic Obstructive/immunology , Sheep , Tissue Kallikreins/immunology , Tissue Kallikreins/metabolism
18.
Clin Cancer Res ; 14(18): 5778-84, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794087

ABSTRACT

PURPOSE: Human tissue kallikreins (KLK) comprise a subgroup of 15 homologous secreted serine proteases. Primarily known for their clinical use as cancer biomarkers (e.g., PSA), KLKs have recently been directly implicated in cancer-related processes, including invasion, angiogenesis, and tumor growth regulation. Therefore, the identification of compounds that would modulate expression of KLKs might be of considerable therapeutic value. EXPERIMENTAL DESIGN: A cell-based high-throughput screening (HTS) of three small molecule libraries ( approximately 4,500 compounds) was undertaken; KLK expression in the breast cancer cell line MDA-MB-468 was assessed with sensitive ELISAs. RESULTS: The initial screening resulted in 66 "putative hits" that decreased KLK5 expression by at least 50% over control. Secondary screening and mini-dose-response assays resulted in 21 "validated hits." These 21 compounds were clustered in only three distinct functional families and were further analyzed in vitro to determine their effectiveness (IC(50)s). Hits that failed to show dose-responsiveness or interfered with the viability of the cells were excluded. Multiple members of the cardiac glycoside family were found to be novel inhibitors of KLK expression, acting at low concentrations (10-50 nmol/L). Furthermore, members of the same family induced marked decreases in c-MYC and c-FOS expression, in a dose-dependent manner that correlated the KLK inhibition, suggesting a transcriptional mechanism of regulation of KLK expression. CONCLUSIONS: We conclude that cardiac glycosides can dramatically suppress the transcription of KLKs and that these effects may be linked to proto-oncogene (c-myc/fos) expression. These findings may partially explain the recently realized antineoplastic actions of cardiac glycosides.


Subject(s)
Breast Neoplasms/metabolism , Cardiac Glycosides/pharmacology , Tissue Kallikreins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Gene Library , Humans , Proto-Oncogene Mas , Transcription, Genetic/drug effects
19.
Biol Chem ; 389(6): 669-80, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18627299

ABSTRACT

Human tissue kallikreins (KLKs) are the largest family of secreted serine protease endopeptidases encoded by 15 genes clustered on chromosome 19q13.4. Multiple KLK enzymes are co-localized in the upper stratum granulosum and stratum corneum of human epidermis, and in associated appendages such as hair follicle epithelia and sweat glands. Until recently, kallikrein proteolytic activity in the skin was exclusively attributed to KLK5 and KLK7. However, wider cutaneous roles of kallikreins became evident in recent years as the proposal of KLK proteolytic activation cascades emerged. We postulate that these proteolytic enzymes may serve as promiscuous mediators of different skin barrier functions, since they are capable of proteolysing different substrates that govern skin desquamation, antimicrobial defense, and lipid permeability. Growing evidence now attests to potential kallikrein involvement in skin inflammation, pigmentation, and tumor suppression via their ability to target proteinase-activated receptor signaling pathways. Current knowledge on kallikrein roles in skin physiology and pathobiology is described in this review.


Subject(s)
Homeostasis , Skin Physiological Phenomena , Skin/metabolism , Tissue Kallikreins/metabolism , Animals , Epidermis/enzymology , Epidermis/metabolism , Gene Expression Regulation, Enzymologic , Humans , Skin/cytology , Skin/enzymology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/genetics
20.
Mol Cancer Res ; 6(6): 1043-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18567807

ABSTRACT

A major characteristic of prostate cancer is the elevation of serum levels of prostate-specific antigen (hK3) and hK2, which are tumor markers that correlate with advancing stages of disease. Including hK4, these three kallikrein serine proteases are almost exclusively produced by the prostate. Prostate cancer cells have been recently shown to overexpress protease-activated receptors (PAR), which can be potentially activated by kallikreins and can regulate tumor growth. Here, we show that recombinant hK2 and hK4 activate ERK1/2 signaling of DU-145, PC-3, and LNCaP prostate cancer cells, which express both PAR1 and PAR2. These kallikreins also stimulate the proliferation of DU-145 cells. Pretreatment of hK2 and hK4 with the serine protease inhibitor, aprotinin, blocks the responses in DU-145 cells, and small interfering RNA against PAR1 and PAR2 also inhibits ERK1/2 signaling. To determine which PAR is activated by hK2 and hK4, a cell line that expresses a single PAR, a PAR1 knockout mouse lung fibroblast cell line transfected with PAR1 (KOLF-PAR1) or PAR2 (KOLF-PAR2) was used. hK4 activates both PAR1 and PAR2, whereas hK2 activates PAR2. hK4 generates more phosphorylated ERK1/2 than hK2. These data indicate that prostatic kallikreins (hK2 and hK4) directly stimulate prostate cancer cell proliferation through PAR1 and/or PAR2 and may be potentially important targets for future drug therapy for prostate cancer.


Subject(s)
Kallikreins/pharmacology , Prostatic Neoplasms/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Tissue Kallikreins/pharmacology , Animals , Aprotinin/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , MAP Kinase Signaling System , Male , Mice , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-2/agonists , Receptor, PAR-2/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...