Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
N Engl J Med ; 390(5): 432-441, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38294975

ABSTRACT

BACKGROUND: Hereditary angioedema is a rare genetic disease that leads to severe and unpredictable swelling attacks. NTLA-2002 is an in vivo gene-editing therapy based on clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9. NTLA-2002 targets the gene encoding kallikrein B1 (KLKB1), with the goal of lifelong control of angioedema attacks after a single dose. METHODS: In this phase 1 dose-escalation portion of a combined phase 1-2 trial of NTLA-2002 in adults with hereditary angioedema, we administered NTLA-2002 at a single dose of 25 mg, 50 mg, or 75 mg. The primary end points were the safety and side-effect profile of NTLA-2002 therapy. Secondary and exploratory end points included pharmacokinetics, pharmacodynamics, and clinical efficacy determined on the basis of investigator-confirmed angioedema attacks. RESULTS: Three patients received 25 mg of NTLA-2002, four received 50 mg, and three received 75 mg. At all dose levels, the most common adverse events were infusion-related reactions and fatigue. No dose-limiting toxic effects, serious adverse events, grade 3 or higher adverse events, or clinically important laboratory findings were observed after the administration of NTLA-2002. Dose-dependent reductions in the total plasma kallikrein protein level were observed between baseline and the latest assessment, with a mean percentage change of -67% in the 25-mg group, -84% in the 50-mg group, and -95% in the 75-mg group. The mean percentage change in the number of angioedema attacks per month between baseline and weeks 1 through 16 (primary observation period) was -91% in the 25-mg group, -97% in the 50-mg group, and -80% in the 75-mg group. Among all the patients, the mean percentage change in the number of angioedema attacks per month from baseline through the latest assessment was -95%. CONCLUSIONS: In this small study, a single dose of NTLA-2002 led to robust, dose-dependent, and durable reductions in total plasma kallikrein levels, and no severe adverse events were observed. In exploratory analyses, reductions in the number of angioedema attacks per month were observed at all dose levels. (Funded by Intellia Therapeutics; ClinicalTrials.gov number, NCT05120830.).


Subject(s)
Angioedemas, Hereditary , CRISPR-Cas Systems , Gene Editing , Adult , Humans , Angioedema , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/genetics , Complement C1 Inhibitor Protein/therapeutic use , Dose-Response Relationship, Drug , Gene Editing/methods , Plasma Kallikrein/genetics , Treatment Outcome
2.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 34(7): 676-681, 2022 Jul.
Article in Chinese | MEDLINE | ID: mdl-36100402

ABSTRACT

OBJECTIVE: To analyze protein profiles in septic patients, and to find potential new targets for the diagnosis and treatment of sepsis. METHODS: A cross sectional observational study was conducted. From January to December 2019, 12 septic patients and 9 healthy volunteers were recruited in the emergency intensive care unit (EICU) of the emergency department of the Affiliated Hospital of Southwest Medical University. The peripheral blood of the two groups was collected for protein mass spectrometry analysis, and the data-independent acquisition technology was used to obtain the expression data of each protein. The obtained data was imported into the online network tool Integrated Differential Expression and Pathway analysis (IDEP2), the data underwent ID converted and were homogenized to verify their comparability, and then principal component analysis was used to eliminate outlier data. Then data with P < 0.05, log2fold change (FC) > 1 or log2FC < -1 were considered to have a statistically significant difference, and the differential proteins were screened out. On the DAVID website, the screened differential proteins would be analyzed by gene ontology (GO), and the biological process, cellular components, and molecular function of the proteins would be analyzed. Protein enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed. Protein-protein interaction (PPI) analysis was performed through the Search Tool for the Retrieval of Interacting Genes Database (STRING) website to find closely related proteins. RESULTS: The data in this study were shown to be comparable after normalization. A total of 125 differential proteins were screened, of which 99 were up-regulated and 26 were down-regulated. GO enrichment analysis discovered that these proteins were mainly extracellular, with cellular regulatory functions and catalytic functions involved in biological regulation, metabolic process and immune process. KEGG pathway analysis suggested that these proteins were involved in amino acid, carbohydrate metabolism and immune-related pathways. PPI analysis showed that key proteins included matrix metalloproteinase 14 (MMP14), fibulin 1 (FBLN1), plasma kallikrein 1 (KLKB1), etc., and finally screened out MMP14 and KLKB1, which were closely related to inflammation and immunity. Both might be potential new targets for early diagnosis and treatment of sepsis. CONCLUSIONS: MMP14 and KLKB1 may be potential biomarkers for the diagnosis, treatment and prognosis of sepsis.


Subject(s)
Computational Biology , Kallikreins/blood , Matrix Metalloproteinase 14/metabolism , Prostate-Specific Antigen/blood , Sepsis , Biomarkers , Computational Biology/methods , Cross-Sectional Studies , Gene Expression Profiling , Gene Regulatory Networks , Humans , Matrix Metalloproteinase 14/genetics , Plasma Kallikrein/genetics , Protein Interaction Maps/genetics , Proteomics , Sepsis/diagnosis , Sepsis/genetics , Sepsis/therapy , Tissue Kallikreins/genetics
3.
Biochem Biophys Res Commun ; 518(3): 409-415, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31451224

ABSTRACT

Particulate matter (PM) is a key component of air pollutants and is associated with mortality of cardiovascular and respiratory diseases. PM-induced tissue injury involves inflammation and coagulation. Plasma prekallikrein (pKal), along with coagulation factor XII (FXII) and high-molecular-weight kininogen (HK), form the plasma kallikrein-kinin system (KKS), a component of the innate immune response that generates proinflammatory products in response to injury. When the KKS proteins contact with activation surface such as negatively charged molecules, this system becomes activated. Activated kallikrein (Kal) activates FXII to initiate the intrinsic coagulation pathway, and cleaves HK to release bradykinin to enhance vascular permeability and systemic inflammation. In his study we determined the role of plasma pKal in the PM2.5-induced lung injury. Using TALEN technology, we generated a new mouse strain lacking the gene for pKal. In PM2.5-induced lung injury model, Klkb1-/- mice exhibited a decrease in total protein, cells numbers in bronchoalveolar lavage fluid (BALF) and histologic lung injury score. The TNF-α and IL-6 levels in BALF were significantly decreased in PM2.5-treated Klkb1-/- mice. Plasma thrombin-antithrombin (TAT) complex levels were significantly decreased in PM2.5-treated Klkb1-/- mice. PM2.5 induces pKal activation, HK cleavage and bradykinin production. PM2.5-induced HK cleavage in plasma was completely blocked by a Kal inhibitor, as well as in pKal-deficient plasma. PM2.5 markedly induced thrombin generation in human plasma and wild-type mouse plasma, which was inhibited by both blockade and deficiency of pKal. Taken together, plasma pKal is activated by PM2.5 and the activated Kal plays an important role in PM2.5-induced lung injury.


Subject(s)
Blood Coagulation , Inflammation/etiology , Lung Injury/etiology , Particulate Matter/adverse effects , Plasma Kallikrein/immunology , Animals , Gene Deletion , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Lung Injury/blood , Lung Injury/genetics , Lung Injury/immunology , Mice , Mice, Knockout , Particulate Matter/immunology , Plasma Kallikrein/analysis , Plasma Kallikrein/genetics
4.
Biochem Biophys Res Commun ; 504(4): 857-864, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30219233

ABSTRACT

Acute liver injury (ALI) is highly lethal acute liver failure caused by different etiologies. Transforming growth factor ß (TGF-ß) is a multifunctional cytokine and a well-recognized inducer of apoptotic and necrotic cell death in hepatocytes. Latent TGF-ß is activated partly through proteolytic cleavage by a serine protease plasma kallikrein (PLK) between the R58 and L59 residues of its propeptide region. Recently, we developed a specific monoclonal antibody to detect the N-terminal side LAP degradation products ending at residue R58 (R58 LAP-DPs) that reflect PLK-dependent TGF-ß activation. This study aimed to explore the potential roles of PLK-dependent TGF-ß activation in the pathogenesis of ALI. We established a mouse ALI model via the injection of anti-Fas antibodies (Jo2) and observed increases in the TGF-ß1 mRNA level, Smad3 phosphorylation, TUNEL-positive apoptotic hepatocytes and R58-positive cells in the liver tissues of Jo2-treated mice. The R58 LAP-DPs were observed in/around F4/80-positive macrophages, while macrophage depletion with clodronate liposomes partly alleviated the Jo2-induced liver injury. Blocking PLK-dependent TGF-ß activation using either the serine proteinase inhibitor FOY305 or the selective PLK inhibitor PKSI-527 or blocking the TGF-ß receptor-mediated signaling pathway using SB431542 significantly prevented Jo2-induced hepatic apoptosis and mortality. Furthermore, similar phenomena were observed in the mouse model of ALI with the administration of acetaminophen (APAP). In summary, R58 LAP-DPs reflecting PLK-dependent TGF-ß activation may serve as a biomarker for ALI, and targeting PLK-dependent TGF-ß activation has potential as a therapeutic strategy for ALI.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Plasma Kallikrein/metabolism , Transforming Growth Factor beta/metabolism , Acetaminophen/adverse effects , Acute Lung Injury/drug therapy , Animals , Antibodies, Monoclonal/adverse effects , Benzamides/pharmacology , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Dioxoles/pharmacology , Disease Models, Animal , Latent TGF-beta Binding Proteins/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Plasma Kallikrein/genetics , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , fas Receptor/immunology
5.
FEBS Lett ; 592(15): 2658-2667, 2018 08.
Article in English | MEDLINE | ID: mdl-30019481

ABSTRACT

Serine proteases play important roles in numerous physiological and pathophysiological processes. Moreover, serine proteases are classical subjects for studies of catalytic and inhibitory mechanisms of enzymes. Here, we determined the crystal structures of a serine protease, murine plasma kallikrein (mPK), and its complex with a peptidic inhibitor. Although mPK in the complex adopts a canonical protease structure, the apo-mPK exhibits a previously unobserved structural feature: the entrance of the intact S1 pocket is blocked by Glu217. In addition, molecular dynamics simulations and functional assays support the flexibility of Glu217 and suggest that this flexibility plays a role in regulating the activity of serine proteases. ENZYMES: EC: 3.4.21.34.


Subject(s)
Catalytic Domain , Plasma Kallikrein/chemistry , Plasma Kallikrein/metabolism , Protein Interaction Domains and Motifs , Amino Acid Sequence/genetics , Animals , Binding Sites/genetics , Catalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Plasma Kallikrein/genetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Substrate Specificity
6.
Acta Physiol (Oxf) ; 224(1): e13060, 2018 09.
Article in English | MEDLINE | ID: mdl-29489063

ABSTRACT

AIM: Recent work has demonstrated that activation of the epithelial sodium channel (ENaC) by aberrantly filtered serine proteases causes sodium retention in nephrotic syndrome. The aim of this study was to elucidate a potential role of plasma kallikrein (PKLK) as a candidate serine protease in this context. METHODS: We analysed PKLK in the urine of patients with chronic kidney disease (CKD, n = 171) and investigated its ability to activate human ENaC expressed in Xenopus laevis oocytes. Moreover, we studied sodium retention in PKLK-deficient mice (klkb1-/- ) with experimental nephrotic syndrome induced by doxorubicin injection. RESULTS: In patients with CKD, we found that PKLK is excreted in the urine up to a concentration of 2 µg mL-1 which was correlated with albuminuria (r = .71) and overhydration as assessed by bioimpedance spectroscopy (r = .44). PKLK increased ENaC-mediated whole-cell currents, which was associated with the appearance of a 67 kDa γ-ENaC cleavage product at the cell surface consistent with proteolytic activation. Mutating a putative prostasin cleavage site in γ-ENaC prevented channel stimulation by PKLK. In a mouse model for nephrotic syndrome, active PKLK was present in nephrotic urine of klkb1+/+ but not of klkb1-/- mice. However, klkb1-/- mice were not protected from ENaC activation and sodium retention compared to nephrotic klkb1+/+ mice. CONCLUSION: Plasma kallikrein is detected in the urine of proteinuric patients and mice and activates ENaC in vitro involving the putative prostasin cleavage site. However, PKLK is not essential for volume retention in nephrotic mice.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney/enzymology , Natriuresis , Nephrotic Syndrome/enzymology , Plasma Kallikrein/metabolism , Water-Electrolyte Balance , Adult , Aged , Animals , Body Composition , Case-Control Studies , Disease Models, Animal , Doxorubicin , Epithelial Sodium Channels/genetics , Female , Humans , Kidney/physiopathology , Male , Membrane Potentials , Mice, Knockout , Middle Aged , Nephrotic Syndrome/genetics , Nephrotic Syndrome/physiopathology , Nephrotic Syndrome/urine , Organism Hydration Status , Plasma Kallikrein/genetics , Plasma Kallikrein/urine , Prospective Studies , Renal Elimination , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/urine , Xenopus laevis
7.
FASEB J ; 31(12): 5419-5431, 2017 12.
Article in English | MEDLINE | ID: mdl-28808141

ABSTRACT

The plasma kallikrein-kinin system (KKS) consists of serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-MW kininogen (HK). Upon activation, activated pKal and FXII cleave HK to release bradykinin. Activation of this system has been noted in patients with rheumatoid arthritis, and its pathogenic role has been characterized in animal arthritic models. In this study, we generated 2 knockout mouse strains that lacked pKal and HK and determined the role of KKS in autoantibody-induced arthritis. In a K/BxN serum transfer-induced arthritis (STIA) model, mice that lacked HK, pKal, or bradykinin receptors displayed protective phenotypes in joint swelling, histologic changes in inflammation, and cytokine production; however, FXII-deficient mice developed normal arthritis. Inhibition of Kal ameliorated arthritis severity and incidence at early stage STIA and reduced the levels of major cytokines in joints. In addition to releasing bradykinin from HK, Kal directly activated monocytes to produce proinflammatory cytokines, up-regulated their C5aR and FcRIII expression, and released C5a. Immune complex increased pKal activity, which led to HK cleavage. The absence of HK is associated with a decrease in joint vasopermeability. Thus, we identify a critical role for Kal in autoantibody-induced arthritis with pleiotropic effects, which suggests that it is a new target for the inhibition of arthritis.-Yang, A., Zhou, J., Wang, B., Dai, J., Colman, R. W., Song, W., Wu, Y. A critical role for plasma kallikrein in the pathogenesis of autoantibody-induced arthritis.


Subject(s)
Arthritis/metabolism , Arthritis/pathology , Autoantibodies/metabolism , Plasma Kallikrein/metabolism , Animals , Arthritis/genetics , Arthritis/immunology , Bradykinin/metabolism , Cytokines/metabolism , Factor XII/genetics , Factor XII/metabolism , Female , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Knockout , Monocytes/metabolism , Plasma Kallikrein/genetics , Polymerase Chain Reaction
8.
Curr Opin Hematol ; 24(5): 411-418, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28604413

ABSTRACT

PURPOSE OF REVIEW: Exposure of blood to foreign surfaces induces reciprocal conversion of the plasma proteins factor XII (fXII) and plasma prekallikrein (PPK) to the proteases α-fXIIa and α-kallikrein. This process, called contact activation, has a range of effects on host defence mechanisms, including promoting coagulation. The nature of the triggering mechanism for contact activation is debated. One hypothesis predicts that fXII has protease activity, either intrinsically or upon surface-binding, that initiates contact activation. We tested this by assessing the proteolytic activity of a recombinant fXII variant that cannot be converted to α-fXIIa. RECENT FINDINGS: The proteolytic activity of fXII-T (for 'triple' mutant), a variant with alanine substitutions for arginine at activation cleavage sites (Arg334, Arg344, and Arg353) was tested with known α-fXIIa substrates. FXII-T activates PPK in solution, and the reaction is enhanced by polyphosphate, an inducer of contact activation released from platelets. In the presence of polyphosphate, fXII-T converts fXII to α-fXIIa, and also converts the coagulation protein factor XI to its active form. SUMMARY: The findings support the hypothesis that contact activation is initiated through activity intrinsic to single-chain fXII, and indicate that preexisting α-fXIIa is not required for induction of contact activation.


Subject(s)
Factor XIIa/metabolism , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Factor XIIa/genetics , Humans , Plasma Kallikrein/genetics , Prekallikrein/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
Blood ; 129(16): 2280-2290, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28130211

ABSTRACT

Thrombolytic therapy using tissue plasminogen activator (tPA) in acute stroke is associated with increased risks of cerebral hemorrhagic transformation and angioedema. Although plasma kallikrein (PKal) has been implicated in contributing to both hematoma expansion and thrombosis in stroke, its role in the complications associated with the therapeutic use of tPA in stroke is not yet available. We investigated the effects of tPA on plasma prekallikrein (PPK) activation and the role of PKal on cerebral outcomes in a murine thrombotic stroke model treated with tPA. We show that tPA increases PKal activity in vitro in both murine and human plasma, via a factor XII (FXII)-dependent mechanism. Intravenous administration of tPA increased circulating PKal activity in mice. In mice with thrombotic occlusion of the middle cerebral artery, tPA administration increased brain hemorrhage transformation, infarct volume, and edema. These adverse effects of tPA were ameliorated in PPK (Klkb1)-deficient and FXII-deficient mice and in wild-type (WT) mice pretreated with a PKal inhibitor prior to tPA. tPA-induced brain hemisphere reperfusion after photothrombolic middle cerebral artery occlusion was increased in Klkb1-/- mice compared with WT mice. In addition, PKal inhibition reduced matrix metalloproteinase-9 activity in brain following stroke and tPA therapy. These data demonstrate that tPA activates PPK in plasma and PKal inhibition reduces cerebral complications associated with tPA-mediated thrombolysis in stroke.


Subject(s)
Angioedema/chemically induced , Cerebral Hemorrhage/chemically induced , Fibrinolytic Agents/adverse effects , Plasma Kallikrein/metabolism , Stroke/drug therapy , Thrombosis/drug therapy , Tissue Plasminogen Activator/adverse effects , Administration, Intravenous , Angioedema/blood , Angioedema/genetics , Animals , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/genetics , Disease Models, Animal , Factor XII/genetics , Factor XII/metabolism , Gene Expression , Humans , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Plasma Kallikrein/genetics , Stroke/blood , Stroke/genetics , Stroke/pathology , Thrombolytic Therapy , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
10.
Ann Neurol ; 77(5): 784-803, 2015 May.
Article in English | MEDLINE | ID: mdl-25628066

ABSTRACT

OBJECTIVE: Recent evidence suggests that ischemic stroke is a thromboinflammatory disease. Plasma kallikrein (PK) cleaves high-molecular-weight kininogen to release bradykinin (BK) and is a key constituent of the proinflammatory contact-kinin system. In addition, PK can activate coagulation factor XII, the origin of the intrinsic coagulation cascade. Thus, PK triggers 2 important pathological pathways of stroke formation, thrombosis and inflammation. METHODS: We investigated the consequences of PK inhibition in transient and permanent models of ischemic stroke. RESULTS: PK-deficient mice of either sex challenged with transient middle cerebral artery occlusion developed significantly smaller brain infarctions and less severe neurological deficits compared with controls without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarctions as well as after permanent stroke. Reduced intracerebral thrombosis and improved cerebral blood flow could be identified as underlying mechanisms. Moreover, blood-brain barrier function was maintained in mice lacking PK, and the local inflammatory response was reduced. PK-deficient mice reconstituted with PK or BK again developed brain infarctions similar to wild-type mice. Important from a translational perspective, inhibition of PK in wild-type mice using a PK-specific antibody was likewise effective even when performed in a therapeutic setting up to 3 hours poststroke. INTERPRETATION: PK drives thrombus formation and inflammation via activation of the intrinsic coagulation cascade and the release of BK but appears to be dispensable for hemostasis. Hence, PK inhibition may offer a safe strategy to combat thromboembolic disorders including ischemic stroke.


Subject(s)
Plasma Kallikrein/metabolism , Stroke/blood , Stroke/prevention & control , Thrombosis/blood , Thrombosis/prevention & control , Animals , Brain Infarction/blood , Brain Infarction/genetics , Brain Infarction/prevention & control , Female , Inflammation/blood , Inflammation/genetics , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Kallikrein/antagonists & inhibitors , Plasma Kallikrein/genetics , Stroke/genetics , Thrombosis/genetics
11.
FASEB J ; 28(2): 923-34, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24249636

ABSTRACT

The soluble cleaved urokinase plasminogen activator receptor (scuPAR) is a circulating protein detected in multiple diseases, including various cancers, cardiovascular disease, and kidney disease, where elevated levels of scuPAR have been associated with worsening prognosis and increased disease aggressiveness. We aimed to identify novel genetic and biomolecular mechanisms regulating scuPAR levels. Elevated serum scuPAR levels were identified in asthma (n=514) and chronic obstructive pulmonary disease (COPD; n=219) cohorts when compared to controls (n=96). In these cohorts, a genome-wide association study of serum scuPAR levels identified a human plasma kallikrein gene (KLKB1) promoter polymorphism (rs4253238) associated with serum scuPAR levels in a control/asthma population (P=1.17 × 10(-7)), which was also observed in a COPD population (combined P=5.04 × 10(-12)). Using a fluorescent assay, we demonstrated that serum KLKB1 enzymatic activity was driven by rs4253238 and is inverse to scuPAR levels. Biochemical analysis identified that KLKB1 cleaves scuPAR and negates scuPAR's effects on primary human bronchial epithelial cells (HBECs) in vitro. Chymotrypsin was used as a proproteolytic control, while basal HBECs were used as a control to define scuPAR-driven effects. In summary, we reveal a novel post-translational regulatory mechanism for scuPAR using a hypothesis-free approach with implications for multiple human diseases.


Subject(s)
Genome-Wide Association Study/methods , Plasma Kallikrein/metabolism , Receptors, Urokinase Plasminogen Activator/blood , Asthma/blood , Binding Sites/genetics , Blotting, Western , Cells, Cultured , Haplotypes , Humans , Linkage Disequilibrium/genetics , Plasma Kallikrein/genetics , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , Pulmonary Disease, Chronic Obstructive/blood , RNA, Messenger/genetics , Receptors, Urokinase Plasminogen Activator/genetics
12.
PLoS One ; 8(7): e68074, 2013.
Article in English | MEDLINE | ID: mdl-23874499

ABSTRACT

Kallikreins are secreted serine proteases with important roles in human physiology. Human plasma kallikrein, encoded by the KLKB1 gene on locus 4q34-35, functions in the blood coagulation pathway, and in regulating blood pressure. The human tissue kallikrein and kallikrein-related peptidases (KLKs) have diverse expression patterns and physiological roles, including cancer-related processes such as cell growth regulation, angiogenesis, invasion, and metastasis. Prostate-specific antigen (PSA), the product of the KLK3 gene, is the most widely used biomarker in clinical practice today. A total of 15 KLKs are encoded by the largest contiguous cluster of protease genes in the human genome (19q13.3-13.4), which makes them ideal for evolutionary analysis of gene duplication events. Previous studies on the evolution of KLKs have traced mammalian homologs as well as a probable early origin of the family in aves, amphibia and reptilia. The aim of this study was to address the evolutionary and functional relationships between tissue KLKs and plasma kallikrein, and to examine the evolution of alternative splicing isoforms. Sequences of plasma and tissue kallikreins and their alternative transcripts were collected from the NCBI and Ensembl databases, and comprehensive phylogenetic analysis was performed by Bayesian as well as maximum likelihood methods. Plasma and tissue kallikreins exhibit high sequence similarity in the trypsin domain (>50%). Phylogenetic analysis indicates an early divergence of KLKB1, which groups closely with plasminogen, chymotrypsin, and complement factor D (CFD), in a monophyletic group distinct from trypsin and the tissue KLKs. Reconstruction of the earliest events leading to the diversification of the tissue KLKs is not well resolved, indicating rapid expansion in mammals. Alternative transcripts of each KLK gene show species-specific divergence, while examination of sequence conservation indicates that many annotated human KLK isoforms are missing the catalytic triad that is crucial for protease activity.


Subject(s)
Alternative Splicing , Evolution, Molecular , Plasma Kallikrein/genetics , Tissue Kallikreins/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Conserved Sequence , Humans , Molecular Sequence Data , Phylogeny , Plasma Kallikrein/chemistry , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Sequence Homology , Tissue Kallikreins/chemistry
13.
Thromb Haemost ; 107(6): 1141-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398951

ABSTRACT

Plasma kallikrein is a multifunctional serine protease involved in contact activation of coagulation. Deficiency in humans is characterised by prolonged activated partial thromboplastin time (aPTT); however, the balance between thrombosis and haemostasis is not fully understood. A study of plasma kallikrein-deficient mice revealed increased aPTT, without prolonged bleeding time. Prekallikrein antisense oligonucleotide (ASO) treatment in mice suggested potential for a positive therapeutic index. The current goal was to further define the role of plasma kallikrein in coagulation. Blood pressure and heart rate were normal in plasma kallikrein-deficient mice, and mice were completely protected from occlusion (100 ± 1.3% control flow) in 3.5% FeCl3 -induced arterial thrombosis versus heterozygotes (20 ± 11.4%) and wild-type littermates (8 ± 0%). Vessels occluded in 8/8 wild-type, 7/8 heterozygotes, and 0/8 knockouts. Anti-thrombotic protection was less pronounced in 5% FeCl3-induced arterial injury. Integrated blood flow was 8 ± 0% control in wild-type and heterozygotes, and significantly (p<0.01) improved to 43 ± 14.2% in knockouts. The number of vessels occluded was similar in all genotypes. Thrombus weight was significantly reduced in knockouts (-47%) and heterozygotes (-23%) versus wild-type in oxidative venous thrombosis. Average tail bleeding time increased modestly in knockout mice compared to wild-type. Average renal bleeding times were similar in all genotypes. These studies confirm and extend studies with prekallikrein ASO, and demonstrate that plasma kallikrein deletion prevents occlusive thrombus formation in mice with a minimal role in provoked bleeding. Additional support for the significance of the intrinsic pathway in the coagulation cascade is provided, as well as for a potential new anti-thrombotic approach.


Subject(s)
Hemostasis , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Thrombosis/prevention & control , Animals , Bleeding Time , Chlorides , Disease Models, Animal , Ferric Compounds , Hemorrhage/blood , Hemorrhage/genetics , Hemostasis/genetics , Heterozygote , Mice , Mice, Knockout , Oligonucleotides, Antisense/metabolism , Partial Thromboplastin Time , Phenotype , Plasma Kallikrein/genetics , Prekallikrein/genetics , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/chemically induced , Venous Thrombosis/genetics , Venous Thrombosis/prevention & control
14.
J Thromb Haemost ; 9(3): 489-95, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21232005

ABSTRACT

BACKGROUND: We evaluated 10 single-nucleotide polymorphisms (SNPs) identified in three European case-control studies as risk factors for venous thrombosis. OBJECTIVES: We sought to replicate the positive findings from this report among Whites and to evaluate the association of these SNPs with venous thrombosis for the first time among Blacks. PATIENT/METHODS: These SNPs were evaluated in a case-control study of deep vein thrombosis and pulmonary embolism that included 1076 cases and 1239 controls. About 50% of subjects were African Americans. We measured plasma factor (F) XI on a subset of subjects. RESULTS: Among Whites, positive findings for rs13146272 in the CYP4V2 gene, for rs3087505 in the KLKB1 gene and for rs3756008 and rs2036914 in the F11 gene were found. We did not find significant associations for rs2227589 in the SERPINC1 gene and for rs1613662 in the GP6 gene. Among Blacks, rs2036914 in F11 and rs670659 in RGS7 were related to venous thrombosis, but the study had limited statistical power for many SNPs. Among Blacks, plasma FXI was related to two SNPs and the OR relating to the 90th percentile of the control distribution of plasma FXI was 2.6 (95% CI, 1.4, 5.0). CONCLUSIONS: Our study supports the finding that genetic variants in the F11 gene are risk factors for venous thrombosis among both Whites and Blacks, although the findings in Blacks require confirmation. A meta-analysis of five case-control studies indicates that rs2227589 in the SERPINC1 gene, rs13146272 in the CYP4V2 gene and rs1613662 in the GP6 gene are risk factors for venous thrombosis among Whites.


Subject(s)
Black or African American/genetics , Polymorphism, Single Nucleotide , Venous Thrombosis/genetics , White People/genetics , Adult , Antithrombin III/genetics , Case-Control Studies , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 4 , Factor XI/genetics , Female , Genetic Predisposition to Disease , Glucose-6-Phosphatase/genetics , Humans , Linkage Disequilibrium , Male , Middle Aged , Plasma Kallikrein/genetics , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Risk Factors , Venous Thrombosis/blood
15.
Neuropeptides ; 44(2): 139-43, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20064660

ABSTRACT

Angiotensin I-converting enzyme (ACE) is recognized as one of the main effector molecules involved in blood pressure regulation. In the last few years some polymorphisms of ACE such as the insertion/deletion (I/D) polymorphism have been described, but their physiologic relevance is poorly understood. In addition, few studies investigated if the specific activity of ACE domain is related to the I/D polymorphism and if it can affect other systems. The aim of this study was to establish a biochemical and functional characterization of the I/D polymorphism and correlate this with the corresponding ACE activity. For this purpose, 119 male brazilian army recruits were genotyped and their ACE plasma activities evaluated from the C- and N-terminal catalytic domains using fluorescence resonance energy transfer (FRET) peptides, specific for the C-domain (Abz-LFK(Dnp)OH), N-domain (Abz-SDK(Dnp)P-OH) and both C- and N-domains (Abz-FRK(Dnp)P-OH). Plasma kallikrein activity was measured using Z-Phe-Arg-AMC as substrate and inhibited by selective plasma kallikrein inhibitor (PKSI). Some physiological parameters previously described related to the I/D polymorphism such as handgrip strength, blood pressure, heart rate and BMI were also evaluated. The genotype distribution was II n=27, ID n=64 and DD n=28. Total plasma ACE activity of both domains in II individuals was significantly lower in comparison to ID and DD. This pattern was also observed for C- and N-domain activities. Difference between ID and DD subjects was observed only with the N-domain specific substrate. Blood pressure, heart rate, handgrip strength and BMI were similar among the genotypes. This polymorphism also affected the plasma kallikrein activity and DD group presents high activity level. Thus, our data demonstrate that the I/D ACE polymorphism affects differently both ACE domains without effects on handgrip strength. Moreover, this polymorphism influences the kallikrein-kinin system of normotensive individuals.


Subject(s)
INDEL Mutation/genetics , Peptidyl-Dipeptidase A/metabolism , Plasma Kallikrein/metabolism , Polymorphism, Genetic/genetics , Analysis of Variance , Blood Pressure/genetics , Fluorescence Resonance Energy Transfer , Genotype , Hand Strength/physiology , Heart Rate/genetics , Humans , Male , Peptidyl-Dipeptidase A/genetics , Plasma Kallikrein/genetics , Young Adult
16.
Gene ; 440(1-2): 57-62, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19344755

ABSTRACT

The transcription start sites (TSSs) of the human plasma prekallikrein gene (KLKB1) determined in RNA from kidney are mostly localized within intron 1 and exon 2, suggesting that the region encompassing exon 1, intron 1, and exon 2 comprises an alternative promoter. Reporter gene analyses in HepG2 and immortalized human kidney epithelial cells confirmed a significant transcriptional activity of this putative promoter region. However, when the TSSs recruited for transcription of the reporter gene were determined, only a few transcripts starting within the insert were detected, whereas the majority of TSSs were located in the vector backbone up to about 2000 bp upstream of the insert. Further reporter gene studies with deletion mutants of the fragment exon 1-intron 1-exon 2 revealed that the 3'-terminal 13-bp segment of intron 1 is sufficient to promote transcriptional activity and induce upstream displacement of the TSS. We conclude that the 13-bp segment represents a cis-acting element which can displace TSSs by provoking the recruitment of alternative promoters and/or by masking intergenic transcription terminating signals.


Subject(s)
Promoter Regions, Genetic/genetics , Transcription Initiation Site , Cell Line , Cells, Cultured , Exons , Humans , Introns , Models, Genetic , Plasma Kallikrein/genetics , Plasma Kallikrein/metabolism , Prekallikrein/genetics , Prekallikrein/metabolism , Transfection
17.
J Biol Chem ; 280(49): 41077-89, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16199530

ABSTRACT

Plasma kallikrein is a serine protease that has many important functions, including modulation of blood pressure, complement activation, and mediation and maintenance of inflammatory responses. Although plasma kallikrein has been purified for 40 years, its structure has not been elucidated. In this report, we described two systems (Pichia pastoris and baculovirus/Sf9 cells) for expression of the protease domain of plasma kallikrein, along with the purification and high resolution crystal structures of the two recombinant forms. In the Pichia pastoris system, the protease domain was expressed as a heterogeneously glycosylated zymogen that was activated by limited trypsin digestion and treated with endoglycosidase H deglycosidase to reduce heterogeneity from the glycosylation. The resulting protein was chromatographically resolved into four components, one of which was crystallized. In the baculovirus/Sf9 system, homogeneous, crystallizable, and nonglycosylated protein was expressed after mutagenizing three asparagines (the glycosylation sites) to glutamates. When assayed against the peptide substrates, pefachrome-PK and oxidized insulin B chain, both forms of the protease domain were found to have catalytic activity similar to that of the full-length protein. Crystallization and x-ray crystal structure determination of both forms have yielded the first three-dimensional views of the catalytic domain of plasma kallikrein. The structures, determined at 1.85 A for the endoglycosidase H-deglycosylated protease domain produced from P. pastoris and at 1.40 A for the mutagenically deglycosylated form produced from Sf9 cells, show that the protease domain adopts a typical chymotrypsin-like serine protease conformation. The structural information provides insights into the biochemical and enzymatic properties of plasma kallikrein and paves the way for structure-based design of protease inhibitors that are selective either for or against plasma kallikrein.


Subject(s)
Gene Expression , Plasma Kallikrein/chemistry , Plasma Kallikrein/genetics , Amino Acid Sequence , Animals , Asparagine/genetics , Baculoviridae/genetics , Binding Sites/genetics , Catalysis , Crystallization , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Glutamic Acid , Glycoside Hydrolases/metabolism , Glycosylation , Humans , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutagenesis , Pichia/genetics , Plasma Kallikrein/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Serine Proteinase Inhibitors/pharmacology , Spodoptera/metabolism , Transfection , Trypsin/metabolism
18.
Biol Reprod ; 70(1): 132-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-13679312

ABSTRACT

Establishment of pregnancy in the pig is accompanied by a localized uterine acute inflammatory response and increase in uterine blood flow. Following rapid trophoblast elongation on Day 12 of pregnancy there is an increase in tissue kallikrein activity and release of bradykinin into the uterine lumen, suggesting the kallikrein-kininogen-kinin system is active in the porcine uterus. The present study investigated endometrial expression and presence of the various factors of the kallikrein-kininogen-kinin system. Endometrial L- and H-kininogen gene expression as well as presence of kininogens in the uterine flushings was evaluated throughout the estrous cycle and early pregnancy in the pig. The possible involvement of plasma kallikrein and Factor XII, activators of the kallikrein-kininogen-kinin system, were evaluated through analysis of gene expression in endometrial and conceptus tissues. Gene expression for plasma kallikrein, Factor XII, and H-kininogen were detected in endometrium but not early conceptus tissues. Factor XII and H-kininogen gene expression were similar across the days of the estrous cycle and early pregnancy. Endometrial plasma kallikrein gene expression was low but increased on Day 15 of the estrous cycle, whereas expression was similar across the days of early pregnancy. In comparison to cyclic gilts, endometrial L-kininogen gene expression increased fourfold on Days 15 and 18 of pregnancy. Both L- and H-kininogen were detected in the uterine flushings of cyclic and pregnant gilts. Presence of L- and H-kininogen in the porcine uterus and endometrial gene expression of plasma kallikrein and Factor XII provide evidence that the kallikrein-kininogen-kinin system is biologically active during establishment of pregnancy in the pig.


Subject(s)
Endometrium/physiology , Factor XII/genetics , Kininogen, High-Molecular-Weight/genetics , Kininogen, Low-Molecular-Weight/genetics , Plasma Kallikrein/genetics , Animals , Blotting, Western , Estrous Cycle/physiology , Female , Gene Expression/physiology , Gestational Age , Liver/physiology , Pregnancy , Swine
19.
Int Immunopharmacol ; 2(13-14): 1867-73, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12489801

ABSTRACT

Plasma prekallikrein (PPK), the zymogen of the contact phase protease plasma kallikrein, forms a non-covalent complex with its substrate H-kininogen (HK). HK binds to cell surface proteoglycans, indirectly anchoring this bradykinin-generating protease to endothelial cells. The heavy chain of PPK consisting of four apple domains designated A1 to A4. Previous studies indicated that a major HK binding site on PPK is within the A2 domain, with additional contributions to binding provided by the N-terminal portion of Al and the central part of A4. To precisely map the relevant binding segments in A2, we employed a monoclonal anti-PPK antibody (PKH6) that binds to A2 and blocks HK-PPK complex formation with an apparent IC50 of 8 nM. Using recombinant A2 C-terminal deletion mutants, we mapped the target epitope of PKH6 to the N-terminal portion of A2, residues 92-153. C-terminal deletion of A2 to residue 145 resulted in a loss of PKH6 binding, as did proteolytic cleavage of A2 at Lys140-Arg141. A comparison of HK binding to various A2 deletion mutants revealed that the major HK binding site is localized to residues 145-153 in the central portion of A2, where it overlaps with the PKH6 epitope. This sequence is conserved in the A2 domain of the related protease factor XI, explaining the unusual strong cross-reactivity of PHK6 with factor XI, as well as the similar HK-binding characteristics of PPK and factor XI.


Subject(s)
Epitopes/metabolism , Kininogen, High-Molecular-Weight/metabolism , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Antibodies, Monoclonal/immunology , Binding Sites , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Epitope Mapping , Epitopes/immunology , Humans , Immunoassay , Plasma Kallikrein/genetics , Plasma Kallikrein/immunology , Prekallikrein/genetics , Prekallikrein/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...