Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Mol Immunol ; 142: 95-104, 2022 02.
Article in English | MEDLINE | ID: mdl-34973499

ABSTRACT

BACKGROUND: Excessive bradykinin (BK) generation from high molecular weight kininogen (HK) by plasma kallikrein (PK) due to lack of protease inhibition is central to the pathophysiology of hereditary angioedema (HAE). Inadequate protease inhibition may contribute to HAE through a number of plasma proteases including factor VII activating protease (FSAP) that can also cleave HK. OBJECTIVE: To investigate the interaction between FSAP and C1 inhibitor (C1Inh) and evaluate the potential role of FSAP in HAE with C1Inh deficiency. MATERIALS AND METHODS: Plasma samples from 20 persons with HAE types 1 or 2 in remission were studied and compared to healthy controls. We measured and compared antigenic FSAP levels, spontaneous FSAP activity, FSAP generation potential, activation of plasma pre-kallikrein (PPK) by FSAP, and the formation of FSAP-C1Inh and FSAP-alpha2-antiplasmin (FSAP-α2AP) complexes. Furthermore, we measured HK cleavage and PK activation after activation of endogenous pro-FSAP and after addition of exogenous FSAP. RESULTS: In plasma from HAE patients, there is increased basal FSAP activity compared to healthy volunteers. HAE plasma exhibits decreased formation of FSAP-C1Inh complexes and increased formation of FSAP-α2AP complexes in histone-activated plasma. Although exogenous FSAP can cleave HK in plasma, this was not seen when endogenous plasma pro-FSAP was activated with histones in either group. PK was also not activated by FSAP in plasma. CONCLUSION: In this study, we established that FSAP activity is increased and the pattern of FSAP-inhibitor complexes is altered in HAE patients. However, we did not find evidence suggesting that FSAP contributes directly to HAE attacks.


Subject(s)
Angioedemas, Hereditary/physiopathology , Complement C1 Inhibitor Protein/genetics , Kininogen, High-Molecular-Weight/metabolism , Serine Endopeptidases/metabolism , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/genetics , Antifibrinolytic Agents/metabolism , Bradykinin/biosynthesis , Factor VII/metabolism , Humans , Kallikreins/blood , Kallikreins/metabolism , Serine Endopeptidases/genetics
2.
Int Immunopharmacol ; 101(Pt A): 108269, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34688137

ABSTRACT

Activated-mast cells (MCs) within gingival-tissue of chronic-periodontitis (CP) patients, release various inflammatory-factors. Bradykinin is a nine-amino-acid peptide and pro-inflammatory mediator, produced through factor-XII-cascade or tryptase-cascade. The ability of MC-chymase in bradykinin generation has not been discussed yet. This study investigated the salivary levels of MC-chymase, high molecular weight kininogen (HMWK) and bradykinin of CP patients; examined the potential of MC-proteases in bradykinin production using biochemistry-models; and explored the effects of bradykinin on gingival fibroblasts (GFs). Saliva-samples were collected; MC-protease activities were detected; HMWK cleavage was assessed by western-blot and SDS-PAGE; bradykinin levels were measured using immunoassay. Primary GFs were extracted and cultured with or without bradykinin; cell-viability, gelatine-zymography and flow-cytometry were applied. Immunocytochemistry and western-blot were used to detect intracellular protein expressions of bradykinin-stimulated GFs. The data showed that the salivary-levels of MC-proteases, bradykinin, HMWK, and lactoferrin of CP-patients were increased. HMWK was cleaved by MC-chymase in-vitro, resulting in bradykinin generation. Bradykinin promoted cell proliferation, cell cycle and matrix-metalloproteinase-2(MMP-2) activity, and increased intracellular expressions of nuclear-factor-kappa-B(NF-κB), focal-adhesion-kinase(FAK), transforming-growth-factor-ß(TGF-ß), P38, P53 of GFs. MC-chymase promotes bradykinin production to stimulate GFs and to continue inflammation during CP development. A new BK-generation cascade found in this study provides a new basis for the pathogenesis of CP and the mechanism of continuous inflammation. The activation of MC-chymase/bradykinin-generation cascade depends on HMWK level and MC-chymase activity under inflammatory condition. MC-chymase contributes to bradykinin production, mediating the cross-talks between MCs and GFs. MC-chymase can be used as a therapeutic target and a salivary biomarker in this case.


Subject(s)
Bradykinin/biosynthesis , Chronic Periodontitis/immunology , Chymases/metabolism , Saliva/chemistry , Adult , Case-Control Studies , Cell Communication/immunology , Cell Cycle/immunology , Cell Proliferation , Chronic Periodontitis/pathology , Chymases/analysis , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Gingiva/cytology , Gingiva/immunology , Gingiva/pathology , Healthy Volunteers , Humans , Kininogen, High-Molecular-Weight/analysis , Lactoferrin/analysis , Male , Mast Cells/enzymology , Mast Cells/immunology , Middle Aged , Saliva/immunology
3.
Postgrad Med ; 133(7): 765-770, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34134576

ABSTRACT

A clinical vignette illustrates a typical presentation of a patient seeking help for acute angioedema. Despite the risks of SARS-CoV-2 (COVID-19) exposure, it is critical to evaluate patients with acute angioedema in person, because there is always the potential for angioedema to progress to the head, neck, or lungs, which can rapidly compromise the airways and require immediate intervention to avoid potential asphyxiation. There are three mediators of angioedema, histamine, leukotriene, or bradykinin, each requiring different management. This article provides clinicians essential information for differentiating between these types of angioedema, including an overview of the underlying pathogenies of angioedema, and the subjective and objective findings that are useful in differentiating between angioedema types. The article ends with the appropriate management for each type of acute angioedema, including the medications approved by the FDA for on-demand treatment of an HAE attack.


Subject(s)
Angioedema/diagnosis , COVID-19/epidemiology , Acute Disease , Angioedema/physiopathology , Angioedema/therapy , Anti-Allergic Agents/therapeutic use , Bradykinin/biosynthesis , Cyclooxygenase 2 Inhibitors/therapeutic use , Diagnosis, Differential , Histamine/biosynthesis , Histamine Antagonists/therapeutic use , Humans , Leukotrienes/biosynthesis , Omalizumab/therapeutic use , Otorhinolaryngologic Surgical Procedures/methods , Physical Examination , SARS-CoV-2
4.
Drugs ; 81(3): 405-409, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33646555

ABSTRACT

Berotralstat (ORLADEYO™) is an orally administered kallikrein inhibitor, which has been developed by BioCryst Pharmaceuticals for hereditary angioedema (HAE). The inhibition of kallikrein by berotralstat decreases the production of bradykinin, which prevents the localised tissue oedema that occurs during attacks of HAE. Berotralstat has been approved in the USA, and subsequently in Japan, for prophylaxis to prevent attacks of HAE in adults and paediatric patients aged 12 years or older. This article summarises the milestones in the development of berotralstat leading to this first approval for prophylaxis to prevent attacks of HAE.


Subject(s)
Angioedemas, Hereditary/drug therapy , Enzyme Inhibitors/pharmacology , Plasma Kallikrein/antagonists & inhibitors , Pyrazoles/pharmacology , Administration, Oral , Angioedemas, Hereditary/metabolism , Bradykinin/antagonists & inhibitors , Bradykinin/biosynthesis , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Plasma Kallikrein/metabolism , Pyrazoles/administration & dosage , Pyrazoles/chemistry
5.
Int J Mol Sci ; 21(9)2020 May 09.
Article in English | MEDLINE | ID: mdl-32397357

ABSTRACT

Recent studies have shown that the innate and adaptive immune system, together with low-grade inflammation, may play an important role in essential hypertension. In this work, to verify the importance of selected factors for the development of essential hypertension, we created a Petri net-based model and analyzed it. The analysis was based mainly on t-invariants, knockouts of selected fragments of the net and its simulations. The blockade of the renin-angiotensin (RAA) system revealed that the most significant effect on the emergence of essential hypertension has RAA activation. This blockade affects: (1) the formation of angiotensin II, (2) inflammatory process (by influencing C-reactive protein (CRP)), (3) the initiation of blood coagulation, (4) bradykinin generation via the kallikrein-kinin system, (5) activation of lymphocytes in hypertension, (6) the participation of TNF alpha in the activation of the acute phase response, and (7) activation of NADPH oxidase-a key enzyme of oxidative stress. On the other hand, we found that the blockade of the activation of the RAA system may not eliminate hypertension that can occur due to disturbances associated with the osmotically independent binding of Na in the interstitium. Moreover, we revealed that inflammation alone is not enough to trigger primary hypertension, but it can coexist with it. We believe that our research may contribute to a better understanding of the pathology of hypertension. It can help identify potential subprocesses, which blocking will allow better control of essential hypertension.


Subject(s)
Essential Hypertension/physiopathology , Inflammation/physiopathology , Models, Biological , Angiotensin II/physiology , Autoantigens/immunology , Blood Coagulation , Bradykinin/biosynthesis , C-Reactive Protein/physiology , Endothelium, Vascular/immunology , Essential Hypertension/etiology , Essential Hypertension/immunology , Humans , Inflammation/immunology , Kallikrein-Kinin System/physiology , Lymphocyte Activation , NADPH Oxidases/physiology , Natriuresis/physiology , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/physiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Skin/physiopathology , Sodium/metabolism , Sodium Chloride, Dietary/pharmacokinetics , Tumor Necrosis Factor-alpha/physiology
6.
Int J Mol Sci ; 21(8)2020 Apr 23.
Article in English | MEDLINE | ID: mdl-32340102

ABSTRACT

The aims of this study were to assess whether ischemic preconditioning (PC) induces bradykinin (Bk) synthesis in bovine aortic endothelial cells (bAECs) and, if so, to explore the molecular mechanisms by which this peptide provides cytoprotection against hypoxia. PC was induced by exposing bAECs to three cycles of 15 min of hypoxia followed by 15 min of reoxygenation. Bk synthesis peaked in correspondence to the early and late phases of PC (10-12 M and 10-11 M, respectively) and was abolished by a selective tissue kallikrein inhibitor, aprotinin. Stimulation with exogenous Bk at concentrations of 10-12 M and 10-11 M reduced the cell death induced by 12 h of hypoxia by 50%. Pretreatment with HOE-140, a Bk receptor 2 (BKR2) inhibitor, in bAECs exposed to 12 h of hypoxia, abrogated the cytoprotective effect of early and late PC, whereas des-Arg-HOE-140, a Bk receptor 1 (BKR1) inhibitor, affected only the late PC. In addition, we found that PC evoked endocytosis and the recycling of BKR2 during both the early and late phases, and that inhibition of these pathways affected PC-mediated cytoprotection. Finally, we evaluated the activation of PKA and Akt in the presence or absence of BKR2 inhibitor. HOE-140 abrogated PKA and Akt activation during both early and late PC. Consistently, BKR2 inhibition abolished cross-talk between PKA and Akt in PC. In bAECs, Bk-synthesis evoked by PC mediates the protection against both apoptotic and necrotic hypoxia-induced cell death in an autocrine manner, by both BKR2- and BKR1-dependent mechanisms.


Subject(s)
Aorta/cytology , Aorta/metabolism , Autocrine Communication , Bradykinin/biosynthesis , Cytoprotection , Endothelial Cells/metabolism , Ischemic Preconditioning , Animals , Apoptosis , Cattle , Endocytosis , Hypoxia/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
7.
Front Immunol ; 10: 2046, 2019.
Article in English | MEDLINE | ID: mdl-31507620

ABSTRACT

Plasminogen activation is essential for fibrinolysis-the breakdown of fibrin polymers in blood clots. Besides this important function, plasminogen activation participates in a wide variety of inflammatory conditions. One of these conditions is hereditary angioedema (HAE), a rare disease with characteristic attacks of aggressive tissue swelling due to unregulated production and activity of the inflammatory mediator bradykinin. Plasmin was already implicated in this disease decades ago, but a series of recent discoveries have made it clear that plasmin actively contributes to this pathology. Collective evidence points toward an axis in which the plasminogen activation system and the contact system (which produces bradykinin) are mechanistically coupled. This is amongst others supported by findings in subtypes of HAE that are caused by gain-of-function mutations in the genes that respectively encode factor XII or plasminogen, as well as clinical experience with the antifibrinolytic agents in HAE. The concept of a link between plasminogen activation and the contact system helps us to explain the inflammatory side effects of fibrinolytic therapy, presenting as angioedema or tissue edema. Furthermore, these observations motivate the development and characterization of therapeutic agents that disconnect plasminogen activation from bradykinin production.


Subject(s)
Bradykinin/biosynthesis , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism , Angioedemas, Hereditary/diagnosis , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/etiology , Angioedemas, Hereditary/metabolism , Animals , Blood Coagulation/drug effects , Brain/metabolism , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/metabolism , Factor XII/metabolism , Humans , Molecular Targeted Therapy
8.
BMC Res Notes ; 12(1): 291, 2019 May 27.
Article in English | MEDLINE | ID: mdl-31133046

ABSTRACT

OBJECTIVE: We recently investigated the pathways of immunoreactive bradykinin (iBK) formation in fresh blood of normal volunteers and of patients with hereditary angioedema due to C1-esterase inhibitor deficiency (HAE-1/-2). Herein, we adapted the techniques to small volumes (200 µl) of previously frozen citrated plasma and further analyzed the mechanisms of iBK formation with additional biotechnological inhibitors. RESULTS: Measurable iBK formation was observed under stimulation with tissue kallikrein (KLK-1, 10 nM), the particulate material Kontact-APTT (concentration reduced to 2% v/v) or recombinant tissue plasminogen activator (tPA, 169 nM), with little background in unstimulated plasma incubated for up to 2 h. Plasma samples from HAE-1/-2 patients responded earlier to tPA than those from controls, as previously reported with whole blood. Lanadelumab inhibited iBK formation induced by Kontact-APTT and tPA. A highly specific plasmin inhibitor, DX-1000, abolished tPA-induced iBK formation in plasma but had no effect against Kontact-APTT, confirming the role of fibrinolysis in tPA-induced kinin formation. The anti-lanadelumab neutralizing antibody M293-D02 reversed the inhibitory effects of lanadelumab. Frozen plasma is a suitable material for measuring iBK formation kinetics, with possible applications such as investigating the effect of rare disease states on the kallikrein-kinin system and monitoring the effect of HAE prophylactic treatments.


Subject(s)
Bradykinin/biosynthesis , Fibrinolysis/physiology , Hereditary Angioedema Types I and II/blood , Kallikreins/chemistry , Tissue Plasminogen Activator/chemistry , Adult , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Neutralizing/chemistry , Antifibrinolytic Agents/chemistry , Blood Specimen Collection/methods , Bradykinin/blood , Case-Control Studies , Female , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Freezing , Humans , Male , Plasma/chemistry , Recombinant Proteins/chemistry
9.
RNA ; 25(2): 255-263, 2019 02.
Article in English | MEDLINE | ID: mdl-30463937

ABSTRACT

Hereditary angioedema (HAE) is a genetic disorder mostly caused by mutations in the C1 esterase inhibitor gene (C1INH) that results in poor control of contact pathway activation and excess bradykinin generation. Bradykinin increases vascular permeability and is ultimately responsible for the episodes of swelling characteristic of HAE. We hypothesized that the use of RNA interference (RNAi) to reduce plasma Factor XII (FXII), which initiates the contact pathway signaling cascade, would reduce contact pathway activation and prevent excessive bradykinin generation. A subcutaneously administered GalNAc-conjugated small-interfering RNA (siRNA) targeting F12 mRNA (ALN-F12) was developed, and potency was evaluated in mice, rats, and cynomolgus monkeys. The effect of FXII reduction by ALN-F12 administration was evaluated in two different vascular leakage mouse models. An ex vivo assay was developed to evaluate the correlation between human plasma FXII levels and high-molecular weight kininogen (HK) cleavage. A single subcutaneous dose of ALN-F12 led to potent, dose-dependent reduction of plasma FXII in mice, rats, and NHP. In cynomolgus monkeys, a single subcutaneous dose of ALN-F12 at 3 mg/kg resulted in >85% reduction of plasma FXII. Administration of ALN-F12 resulted in dose-dependent reduction of vascular permeability in two different mouse models of bradykinin-driven vascular leakage, demonstrating that RNAi-mediated reduction of FXII can potentially mitigate excess bradykinin stimulation. Lastly, ex vivo human plasma HK cleavage assay indicated FXII-dependent bradykinin generation. Together, these data suggest that RNAi-mediated knockdown of FXII by ALN-F12 is a potentially promising approach for the prophylactic treatment of HAE.


Subject(s)
Angioedemas, Hereditary/drug therapy , Bradykinin/biosynthesis , Factor XII/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Animals , Capillary Permeability/drug effects , Complement C1 Inhibitor Protein/genetics , Factor XII/analysis , Female , Humans , Kininogens/metabolism , Macaca fascicularis , Mice , Mice, Inbred C57BL , RNA Interference , Rats
10.
J Thromb Haemost ; 16(9): 1674-1685, 2018 09.
Article in English | MEDLINE | ID: mdl-29920929

ABSTRACT

The plasma contact system contributes to thrombosis in experimental models. Even though our standard blood coagulation tests are prolonged when plasma lacks contact factors, this enzyme system appears to have a minor (if any) role in hemostasis. In this review, we explore the clinical phenotype of C1 esterase inhibitor (C1-INH) deficiency. C1-INH is the key plasma inhibitor of the contact system enzymes, and its deficiency causes hereditary angioedema (HAE). This inflammatory disorder is characterized by recurrent aggressive attacks of tissue swelling that occur at unpredictable locations throughout the body. Bradykinin, which is considered to be a byproduct of the plasma contact system during in vitro coagulation, is the main disease mediator in HAE. Surprisingly, there is little evidence for thrombotic events in HAE patients, suggesting mechanistic uncoupling from the intrinsic pathway of coagulation. In addition, it is questionable whether a surface is responsible for contact system activation in HAE. In this review, we discuss the clinical phenotype, disease modifiers and diagnostic challenges of HAE. We subsequently describe the underlying biochemical mechanisms and contributing disease mediators. Furthermore, we review three types of HAE that are not caused by C1-INH inhibitor deficiency. Finally, we propose a central enzymatic axis that we hypothesize to be responsible for bradykinin production in health and disease.


Subject(s)
Angioedemas, Hereditary/blood , Blood Coagulation/physiology , Bradykinin/physiology , Age of Onset , Angioedemas, Hereditary/enzymology , Angioedemas, Hereditary/etiology , Angioedemas, Hereditary/physiopathology , Bradykinin/biosynthesis , Capillary Permeability , Complement Activation , Complement C1 Inhibitor Protein/physiology , Factor XIIa/physiology , Female , Hereditary Angioedema Types I and II/blood , Hereditary Angioedema Types I and II/enzymology , Hereditary Angioedema Types I and II/physiopathology , Humans , Inflammation , Kallidin/metabolism , Kallikreins/physiology , Kininogen, High-Molecular-Weight/metabolism , Male , Models, Biological , Phenotype , Polyphosphates/metabolism , Serine Proteinase Inhibitors/deficiency , Serine Proteinase Inhibitors/physiology
11.
Ann Allergy Asthma Immunol ; 114(3): 245-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25601538

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme inhibitor-induced angioedema (ACEI-AE) is mediated by bradykinin. There remains an unmet treatment need because these patients, when presenting to the emergency department (ED), do not respond to conventional therapies, such as antihistamines and corticosteroids. OBJECTIVE: To estimate the treatment effect of ecallantide, a recombinant plasma kallikrein inhibitor, in ED patients with ACEI-AE in whom conventional therapy fails. METHODS: This was a triple-blind (patient, physician, and statistician), randomized, controlled, phase 2 study to estimate the magnitude of safety and efficacy signals for designing a definitive phase 3 trial comparing conventional therapy with ecallantide to conventional therapy with placebo. Patients were enrolled from April 1, 2010, through January 31, 2013. The primary efficacy study end point was achieving discharge criteria from the ED within 4 hours after initiating study-related treatment. RESULTS: Discharge criteria from the ED was met in 4 hours or less for 8 (31%) of 26 patients receiving ecallantide vs 5 of (21%) 24 patients receiving placebo (difference in proportions, 10%; 95% confidence interval, -14% to 34%). Ecallantide was well tolerated in both groups. CONCLUSION: The results from this preliminary study reveal that ecallantide is safe to use and may increase the proportion of patients who meet early discharge criteria by approximately10%. A larger phase 3 study is necessary to confirm the efficacy and evaluate the cost-effectiveness of ecallantide use for ACEI-AE in the ED setting. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01036659.


Subject(s)
Angioedema/drug therapy , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Peptides/therapeutic use , Angioedema/chemically induced , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Bradykinin/biosynthesis , Double-Blind Method , Emergency Service, Hospital , Female , Humans , Kallikreins/antagonists & inhibitors , Male , Middle Aged , Peptides/adverse effects , Placebos/administration & dosage
12.
Allergy ; 70(1): 115-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25186184

ABSTRACT

BACKGROUND: Hereditary angioedema types I and II are caused by a functional deficiency of C1 inhibitor (C1-INH), leading to overproduction of bradykinin. The current functional diagnostic assays employ inhibition of activated C1s; however, an alternative, more physiologic method is desirable. METHODS: ELISAs were developed using biotinylated activated factor XII (factor XIIa) or biotinylated kallikrein bound to avidin-coated plates. Incubation with plasma was followed by detection of bound C1-INH. RESULTS: After standard curves were developed for quantification of C1-INH, serial dilutions of normal plasma were employed to validate the ability to detect known concentration of C1-INH in the plasma as a percent of normal. Hereditary angioedema (HAE) types I and II were then tested. The level of functional C1-INH in all HAE types I and II plasma tested was less than 40% of our normal control. This was evident regardless of whether we measured factor XIIa-C1-INH or kallikrein-C1-INH complexes, and the two assays were in close agreement. By contrast, testing the same samples utilizing the commercial method (complex ELISA, Quidel Corp.) revealed the levels of C1-INH between 0 and 57% of normal (mean, 38%), and 42 samples were considered equivocal (four controls and 38 patients). CONCLUSIONS: Diagnosis of HAE types I and II can be ascertained by inhibition of enzymes of the bradykinin-forming cascade, namely factor XIIa and kallikrein. Either method yields functional C1-INH levels in patients with HAE (types I and II) that are clearly abnormal with less variance or uncertainty than the commercial method.


Subject(s)
Angioedemas, Hereditary/diagnosis , Bradykinin/biosynthesis , Factor XIIa , Plasma Kallikrein , Angioedemas, Hereditary/enzymology , Case-Control Studies , Complement C1 Inhibitor Protein/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Reproducibility of Results , Sensitivity and Specificity
13.
J Clin Apher ; 29(2): 90-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24023037

ABSTRACT

We evaluated the bradykinin generation level during leukocytapheresis (LCAP) using novel Cellsorba(TM) CS-180S, which has sodium pyrosulfite and sodium carbonate as a filling solution. Subjects of this study were 14 rheumatoid arthritis patients. Regardless of the type of anticoagulant used, bradykinin levels were lower with the novel CS-180S than with the conventional CS-180S (28.7 ± 53.3 vs. 8.0 ± 2.7 as the mean ± standard deviation). When anticoagulants other than nafamostat mesilate were used with the conventional CS-180S, bradykinin levels increased at the column outlet compared with the column inlet, and adverse effects of bradykinin were seen in several cases. In contrast, bradykinin levels remained low and no bradykinin-associated adverse events were observed with the novel CS-180S. We recommend using the novel column instead of the conventional column in the treatment of LCAP.


Subject(s)
Bradykinin/biosynthesis , Leukapheresis/methods , Blood Coagulation , Bradykinin/adverse effects , Humans , Solutions
15.
Clin Exp Pharmacol Physiol ; 38(9): 623-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21736602

ABSTRACT

1. Aliskiren is a renin inhibitor with an IC(50) of 0.6 nmol/L for human renin, 4.5 nmol/L for mouse renin and 80 nmol/L for rat renin. 2. In the present study, we compared the effects of aliskiren (10 mg/kg per day), the angiotensin-converting enzyme inhibitor perindopril (0.2 mg/kg per day) and their combination on angiotensin and bradykinin peptides in female heterozygous (mRen-2)27 rats, transgenic for the mouse renin gene. 3. All three treatments produced similar reductions in systolic blood pressure, heart weight and plasma aldosterone levels and reduced angiotensin II levels in lung, but only perindopril and the combination reduced angiotensin II levels in kidney of (mRen-2)27 rats. In contrast, aliskiren and the combination, but not perindopril alone, increased cardiac bradykinin levels. Aliskiren increased immunostaining for tissue kallikrein in the heart and reduced cardiac fibrosis. 4. We investigated the mechanism underlying the increase in bradykinin levels following aliskiren treatment in Sprague-Dawley rats, in which aliskiren has a lower potency for renin inhibition. Aliskiren (10 mg/kg per day) reduced renal angiotensin levels within 24 h, but treatment for > 24 h was required to increase cardiac bradykinin levels. Moreover, 3 mg/kg per day aliskiren increased cardiac bradykinin levels, but did not reduce renal angiotensin levels. Aliskiren did not potentiate the hypotensive effects of bradykinin; however, it increased tissue kallikrein, but not plasma kallikrein, mRNA levels in the heart. 5. These data demonstrate that the aliskiren-induced increase in cardiac bradykinin levels is independent of renin inhibition and changes in bradykinin metabolism, but is associated with increased tissue kallikrein gene expression.


Subject(s)
Amides/pharmacology , Bradykinin/genetics , Fumarates/pharmacology , Heart/drug effects , RNA, Messenger/biosynthesis , Tissue Kallikreins/genetics , Aldosterone/blood , Angiotensin II/antagonists & inhibitors , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Bradykinin/biosynthesis , Female , Kallikreins/genetics , Kallikreins/metabolism , Kidney/drug effects , Kidney/metabolism , Lung/drug effects , Lung/metabolism , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Perindopril/pharmacology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Renin/antagonists & inhibitors , Renin/genetics , Tissue Kallikreins/biosynthesis
16.
Immunity ; 34(2): 258-68, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21349432

ABSTRACT

Activated mast cells trigger edema in allergic and inflammatory disease. We report a paracrine mechanism by which mast cell-released heparin increases vascular permeability in vivo. Heparin activated the protease factor XII, which initiates bradykinin formation in plasma. Targeting factor XII or kinin B2 receptors abolished heparin-triggered leukocyte-endothelium adhesion and interfered with a mast cell-driven drop in blood pressure in rodents. Intravital laser scanning microscopy and tracer measurements showed heparin-driven fluid extravasation in mouse skin microvessels. Ablation of factor XII or kinin B2 receptors abolished heparin-induced skin edema and protected mice from allergen-activated mast cell-driven leakage. In contrast, heparin and activated mast cells induced excessive edema in mice deficient in the major inhibitor of factor XII, C1 esterase inhibitor. Allergen exposure triggered edema attacks in hereditary angioedema patients, lacking C1 esterase inhibitor. The data indicate that heparin-initiated bradykinin formation plays a fundamental role in mast cell-mediated diseases.


Subject(s)
Bradykinin/biosynthesis , Capillary Leak Syndrome/physiopathology , Capillary Permeability/physiology , Heparin/physiology , Mast Cells/metabolism , Passive Cutaneous Anaphylaxis/physiology , Animals , Bradykinin/genetics , Capillary Leak Syndrome/etiology , Cell Adhesion , Complement C1 Inhibitor Protein/physiology , Edema/etiology , Edema/physiopathology , Endothelial Cells/pathology , Enzyme Activation , Factor XII/physiology , Heparin/metabolism , Hypotension/etiology , Hypotension/physiopathology , Immunoglobulin E/immunology , Kallikrein-Kinin System/physiology , Leukocytes/physiology , Male , Mice , Paracrine Communication/physiology , Plasma , Rats , Signal Transduction/physiology , Skin/blood supply
17.
Biochem Biophys Res Commun ; 405(3): 338-43, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21167814

ABSTRACT

We have previously cloned a cDNA encoding human prolylcarboxypeptidase (PRCP) and expressed the cDNA in the Schneider 2 (S2) drosophila cell line. Here, we further characterized this recombinant enzyme. Investigations were performed to determine whether recombinant PRCP (rPRCP) metabolizes kinins (BK 1-9 and BK 1-8). The metabolites of these kinins were identified by LC/MS. rPRCP metabolized BK 1-8 to BK 1-7, whereas rPRCP was ineffective in metabolizing BK 1-9. The hydrolysis of BK 1-8 by rPRCP was dose- and time-dependent. A homology model of PRCP was developed based upon the sequence of dipeptidyl-peptidase 7 (DPP7, PDB ID: 3JYH), and providentially, the structure of PRCP (PDB ID: 3N2Z) was characterized during the course of our investigation. Docking studies of bradykinin oligopeptides were performed both from the homology model, and from the crystal structure of PRCP. These docking studies may provide a better understanding of the contribution of specific residues involved in substrate selectivity of human PRCP.


Subject(s)
Carboxypeptidases/metabolism , Kinins/metabolism , Animals , Bradykinin/biosynthesis , Bradykinin/chemistry , Carboxypeptidases/chemistry , Carboxypeptidases/genetics , Catalytic Domain , Chromatography, Liquid , DNA, Complementary/genetics , Drosophila , Humans , Hydrogen Bonding , Hydrolysis , Kinins/chemistry , Mass Spectrometry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
18.
Australas J Dermatol ; 51(3): 157-62, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695852

ABSTRACT

Hereditary angio-oedema (HAE) is a rare but potentially life-threatening condition. Three types are now recognized. Types I and II HAE involve mutations in the C1NH (SERPING1) gene, encoding the C1 inhibitor protein, whereas type III HAE involves mutations in the F12 gene, encoding coagulation factor XII (Hageman factor). They share a common final pathway leading to increased bradykinin formation. HAE must be distinguished from acquired angio-oedema with C1 esterase inhibitor deficiency, angiotensin-converting enzyme inhibitor-induced angio-oedema and the much more common histaminergic angio-oedema, occurring with or without weals. Understanding the pathogenesis of HAE is leading to the introduction of new therapies that target the bradykinin receptor or inhibit kallikrein activity, innovations that will hopefully reduce morbidity and mortality in this group of severe genetic disease.


Subject(s)
Angioedemas, Hereditary/diagnosis , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/classification , Angioedemas, Hereditary/genetics , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Bradykinin/biosynthesis , Bradykinin Receptor Antagonists , Complement C1 Inactivator Proteins/genetics , Complement C1 Inhibitor Protein , Estrogens/adverse effects , Factor XII/genetics , Female , Humans , Kallikreins/antagonists & inhibitors , Mutation
19.
Mol Immunol ; 47(13): 2161-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20580091

ABSTRACT

The plasma bradykinin-forming cascade and the complement pathways share many elements, including cross-activation, common control mechanisms, and shared binding proteins. The C1 inhibitor (C1 INH) is not only the inhibitor of activated C1r and C1s, but it is the key control protein of the plasma bradykinin-forming cascade. It inhibits the autoactivation of Factor XII, the ability of Factor XIIa to activate prekallikrein and Factor XI, the activation of high molecular weight kininogen (HK) by kallikrein, and the feedback activation of Factor XII by kallikrein. Thus in the absence of C1 INH (hereditary angioedema or acquired C1 INH deficiency) there is unimpeded formation of bradykinin leading to angioedema. Activated Factor XII (Factor XIIa, 80,000 kDa) is further cleaved by kallikrein or plasmin to yield Factor XII fragment (Factor XIIf, 30,000 kDa) and Factor XIIf can activate the C1r subcomponent of C1, particularly when C1 INH (which inhibits Factor XIIf) is absent. Once bradykinin is formed, it causes vasodilatation and increased vascular permeability by interaction with constitutively expressed B-2 receptors. However degradation of bradykinin by carboxypeptidase N (in plasma) or carboxypeptidase M (on endothelial cells) yields des-arg-9 (Kerbiriou and Griffin, 1979) bradykinin which interacts with B-1 receptors. B-1 receptors are induced in inflammatory states by cytokines such as Interleukin 1 and its interaction with bradykinin may prolong or perpetuate the vascular response until bradykinin is completely inactivated by angiotensin converting enzyme or aminopeptidase P, or neutral endopeptidase. The entire bradykinin-forming cascade is assembled and can be activated along the surface of endothelial cells in zinc dependent reactions involving gC1qR, cytokeratin 1, and the urokinase plasminogen activated receptor (u-PAR). Although Factors XII and HK can be shown to bind to each one of these proteins, they exist in endothelial cells as two bimolecular complexes; gC1qR-cytokeratin 1, which preferentially binds HK, and cytokeratin 1-u-PAR which preferentially binds Factor XII. The gC1qR, which binds the globular heads of C1q is present in excess and can bind either Factor XII or HK however the binding sites for HK and C1q have been shown to reside at opposite ends of gC1qR. Activation of the bradykinin-forming pathway can be initiated at the cell surface by gC1qR-induced autoactivation of Factor XII or direct activation of the prekallikrein-HK complex by endothelial cell-derived heat-shock protein 90 (HSP 90) or prolylcarboxypeptidase with recruitment or Factor XII by the kallikrein produced.


Subject(s)
Biosynthetic Pathways , Bradykinin/biosynthesis , Complement Activation/immunology , Complement System Proteins/immunology , Animals , Bradykinin/blood , Complement C1 Inhibitor Protein/metabolism , Complement System Proteins/metabolism , Factor XII/metabolism , Humans , Kallikreins/metabolism , Kininogen, High-Molecular-Weight/metabolism
20.
Ann Allergy Asthma Immunol ; 104(1): 50-4, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20143645

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) is typically the result of a deficiency of C1 inhibitor (C1-INH) with gene defects that lead to diminished plasma levels or the production of a dysfunctional protein. Replacement therapy with C1-INH has been shown to be effective in ameliorating episodes of swelling. We have reported elevated baseline levels of bradykinin, C4a, and plasmin-alpha2-antiplasmin complexes in the plasma of patients with HAE compared with the plasma of healthy controls. The production of factor XII fragment on in vitro activation of plasma with HAE has also been observed. OBJECTIVE: To perform serial assessment of abnormalities of the bradykinin-forming pathway and fibrinolysis in patients with HAE after treatment of episodes of swelling with intravenous C1-INH. METHODS: We obtained samples of plasma from 9 patients with HAE at a quiescent period (baseline), during an attack of swelling, and at 1, 4, and 12 hours after termination of an infusion of C1-INH. Factor XIIa, kallikrein, and plasmin were each measured by cleavage of synthetic substrates specific for each item. RESULTS: Each enzyme was strikingly elevated at baseline compared with the levels in pooled healthy plasma, and there was a progressive decline of activity to normal for factor XIIa and plasmin. Kallikrein decreased in 7 of the 9 patients at 1 hour and then decreased in all patients. Bradykinin levels were elevated at the outset in all patients, increased prominently during an attack of swelling, decreased to baseline after 1 hour, and then decreased toward normal by 4 and 12 hours. CONCLUSION: The plasma levels of factor XIIa, kallikrein, and bradykinin decreased when measured serially subsequent to the infusion of nanofiltered C1-INH.


Subject(s)
Angioedemas, Hereditary/drug therapy , Bradykinin/biosynthesis , Complement C1 Inhibitor Protein/metabolism , Complement C1 Inhibitor Protein/pharmacology , Fibrinolysis/drug effects , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/genetics , Angioedemas, Hereditary/physiopathology , Bradykinin/antagonists & inhibitors , Bradykinin/blood , Bradykinin/genetics , Complement Activation/drug effects , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/immunology , Complement C4a/metabolism , Factor XIIa/metabolism , Fibrinolysin/metabolism , Fibrinolysin/pharmacology , Fibrinolysis/genetics , Fibrinolysis/immunology , Humans , Infusions, Intravenous , Kallikreins/blood , Kallikreins/pharmacology , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...