Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Allergy ; 78(5): 1333-1346, 2023 05.
Article in English | MEDLINE | ID: mdl-36789476

ABSTRACT

BACKGROUND: Over-release of the vasoactive peptide bradykinin (BK) due to mutation in the SERPING1 gene is the leading cause of hereditary angioedema (HAE). BK directly activates endothelial cells and increases vascular permeability by disrupting the endothelial barrier, leading to angioedema affecting face, lips, extremities, gastrointestinal tract, and larynx. Although various pharmacological treatment options for HAE became available during the last decade, they are presently limited and pose a major economic burden on patients. To identify additional therapeutic options for HAE, we evaluated the effect of CU06-1004, an endothelial dysfunction blocker, on BK-induced vascular hyperpermeability and the HAE murine model. METHODS: To investigate the effect of CU06-1004 on BK-induced vascular hyperpermeability in vivo, we pre-administrated WT mice with the drug and then induced vascular leakage through intravenous injection of BK and observed vascular alternation. Then, SERPING1 deficient mice were used for a HAE murine model. For an in vitro model, the HUVEC monolayer was pre-treated with CU06-1004 and then stimulated with BK. RESULTS: Bradykinin disrupted the endothelial barrier and formed interendothelial cell gaps, leading to hyperpermeability in vivo and in vitro. However, CU06-1004 treatment protected the endothelial barrier by suppressing Src and myosin light chain activation via BK and alleviated hyperpermeability. CONCLUSION: Our study shows that CU06-1004 oral administration significantly reduced vascular hyperpermeability in the HAE murine model by protecting the endothelial barrier function against BK stimulation. Therefore, protecting endothelium against BK with CU06-1004 could serve as a potential prophylactic/therapeutic approach for HAE patients.


Subject(s)
Angioedemas, Hereditary , Animals , Mice , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/genetics , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/pharmacology , Bradykinin/pharmacology , Endothelial Cells , Disease Models, Animal , Endothelium
2.
Am J Emerg Med ; 49: 6-9, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34029784

ABSTRACT

INTRODUCTION: Angiotensin-converting enzyme inhibitor (ACEi)-induced angioedema is a serious emergency that can cause life-threatening symptoms and death if not treated promptly. Potential treatment options for ACEi-induced angioedema include medications with limited evidence for use in this patient population. The purpose of this study was to evaluate the use, clinical efficacy, and angioedema-related medication costs of C1 esterase inhibitor (C1EI) for ACEi-induced angioedema. METHODS: This was a retrospective, propensity-matched cohort study comparing patients who received C1EI to those who did not receive C1EI for ACEi-induced angioedema. The primary outcome of interest was comparing the proportion of patients who required intubation secondary to ACEi-induced angioedema. Secondary endpoints of interest were also included. RESULTS: After propensity score matching, 22 patients were stratified into both the non-C1EI group and C1EI group, respectively. There was no difference between the groups with respect to the proportion of intubation (13.6% in the C1EI group vs. 9.1% in the non-C1EI group, p > 0.999). Mean cost of angioedema-related medication therapy was higher in the C1EI group compared to the non-C1EI group [$8758.95 (± $2959.30) vs. $15.91 (± $7.32), p < 0.001]. CONCLUSIONS: In this retrospective cohort study, the use of C1EI for ACEi-induced angioedema did not demonstrate improved outcomes with respect to intubation and resulted in increased costs. Larger, multicenter, prospective studies are needed to further validate the results of this study and to provide more clarity on the role of C1EI therapy in ACEi-induced angioedema.


Subject(s)
Angioedema/etiology , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Complement C1 Inhibitor Protein/pharmacology , Aged , Angioedema/physiopathology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cohort Studies , Complement C1 Inhibitor Protein/therapeutic use , Female , Humans , Male , Middle Aged , Propensity Score , Prospective Studies , Retrospective Studies
3.
Int Arch Allergy Immunol ; 179(2): 158-164, 2019.
Article in English | MEDLINE | ID: mdl-30893690

ABSTRACT

BACKGROUND: We have previously shown that human monocyte-derived dendritic cells (moDCs) may participate in immune system-mediated hypercoagulable state through enhanced tissue factor (TF) expression and that the complement system may be involved in this process. OBJECTIVES: The aim of this study was to explore the role of pentraxin 3 (PTX3) and the complement system in enhanced TF expression in moDCs. METHODS: moDCs were generated from isolated human monocytes. PTX3 levels in whole human blood supplemented with moDCs were determined after lipopolysaccharide (LPS) stimulation. PTX3 release by the generated moDCs upon LPS stimulation was also assessed. The effect of PTX3 on whole blood coagulation was investigated using thromboelastometric analysis. TF expression in stationary moDCs treated with LPS and/or PTX3 was determined by measuring TF activity. The effect of complement inhibitors on TF activity in moDCs treated with LPS and/or PTX3 under low-shear conditions was evaluated. RESULTS: PTX3 levels were higher in whole blood supplemented with moDCs than in the presence of monocytes and were further elevated by LPS stimulation. PTX3 release from generated moDCs was also increased by LPS stimulation. PTX3 reduced whole blood coagulation time in a dose-dependent manner. However, PTX3 did not increase TF expression in stationary moDCs. Under low-shear conditions, PTX3 increased TF expression in moDCs. C1 esterase inhibitor (C1-inh) suppressed this effect. CONCLUSIONS: PTX3 might have a thrombophilic activity and enhance TF expression in moDCs under low-shear conditions. Furthermore, suppression of moDC-associated hypercoagulability by C1-inh might be partly ascribed to its inhibitory effect on PTX3.


Subject(s)
C-Reactive Protein/metabolism , Complement C1 Inhibitor Protein/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Serum Amyloid P-Component/metabolism , Thromboplastin/genetics , Adult , Blood Coagulation , Enzyme Activation , Female , Humans , Lipopolysaccharides/immunology , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Shear Strength , Thrombelastography
4.
BMC Vet Res ; 15(1): 475, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888626

ABSTRACT

BACKGROUND: Immune-mediated hemolytic anemia (IMHA) is a common disease that affects all breeds of dogs and is associated with significant morbidity and mortality. Intravascular hemolysis of erythrocytes in IMHA is caused by complement activation and is often fatal. No current treatments target complement activation in canine IMHA. Human C1 esterase (C1-INH) reduces canine complement-mediated hemolysis in vitro, and a recent pharmacokinetic analysis of an FDA licensed formulation of C1-INH in dogs confirmed that a 50 IU/kg dose of C1-INH is safe to administer to dogs, and effectively inhibits canine complement mediated hemolysis ex-vivo. The C1INCH randomized controlled trial will evaluate the efficacy of this drug in dogs with intravascular hemolysis. METHODS: We will conduct a multicenter, placebo-controlled double-blind randomized clinical trial of C1-INH in dogs with intravascular hemolysis due to IMHA. We will randomize 18 dogs to receive three doses of intravenous C1-INH or saline in 24 h. Immunosuppressive and antithrombotic therapies will be standardized. Primary outcome measures will be changes in plasma free hemoglobin, serum concentrations of LDH, bilirubin, and haptoglobin. Using patient samples, we will evaluate complement activation in canine IMHA using a novel C5b-9 ELISA assay, flow cytometric detection of C3b on RBC, and by measurement of residual plasma complement activity. Secondary outcome measures will be survival to hospital discharge, duration of hospitalization, number and volume of red blood cell transfusions, and rescue therapy requirements. We will monitor dogs for adverse drug reactions. Sample size was estimated from pilot data on LDH and hemolysis index (HI) in dogs with IMHA. To detect 2-way differences between the upper and lower 50% of the LDH and HI values of equivalent size with 80% power at P < 0.05 will require 9 dogs in each arm. DISCUSSION: We anticipate that IV administration of C1-INH will significantly inhibit complement mediated hemolysis in dogs with intravascular IMHA, as determined by blood biomarker measurements (decreased plasma hemoglobin, LDH and bilirubin, increased haptoglobin). We expect this will translate into significant reductions in transfusion requirements and duration of hospitalization. TRIAL REGISTRATION: This trial has been prospectively registered with the AVMA registry (AAHSD005025).


Subject(s)
Anemia, Hemolytic, Autoimmune/veterinary , Complement C1 Inhibitor Protein/pharmacology , Dog Diseases/drug therapy , Anemia, Hemolytic, Autoimmune/drug therapy , Animals , Dogs , Female , Hemolysis/drug effects , Male , Randomized Controlled Trials as Topic
5.
J Thromb Thrombolysis ; 48(1): 81-87, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31030323

ABSTRACT

C1-inhibitor (C1INH) was shown to enhance thrombin generation (TG) in the presence of thrombomodulin (TM) by reducing production of activated protein C. Because C1INH is known to inhibit fibrinolytic system proteases, the objective of this study was to evaluate the effect of moderate (3 IU/ml) and high (16 IU/ml) C1INH concentrations on TG and plasmin generation (PG) in the presence of TM. These concentrations were evaluated based on expected maximum plasma levels following C1INH replacement therapy and recently suggested supraphysiologic dosing. TG and PG were investigated in platelet poor plasmas obtained from 21 healthy donors. An assay designed to monitor the continuous generation of the 7-amino-4-methylcoumarin fluorescence from substrates specific to thrombin or plasmin was used to evaluate the impact of C1INH activity. To characterize the C1INH effects on TG and PG, the thrombin and plasmin concentration peaks and production rates were calculated. TM addition to donor plasma shifted the concentration dependence of C1INH on TG parameters from reduction to enhancement. Conversely, PG parameters were significantly reduced by 16 IU/ml in both the presence and absence of TM. Moderate C1INH concentration (3 IU/ml) reduced TG and PG in the absence of TM but did not significantly affect these parameters in the presence of TM. Finally, 3 IU/ml of C1INH reduced PG more so than TG in the absence of TM. The presented results suggest a mechanism by which C1INH could potentiate thrombosis by inhibition of fibrinolysis.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Fibrinolysin/antagonists & inhibitors , Thrombin/drug effects , Thrombomodulin/physiology , Blood Coagulation , Blood Specimen Collection , Dose-Response Relationship, Drug , Fibrinolysin/biosynthesis , Fibrinolysis/drug effects , Healthy Volunteers , Humans , Thrombin/metabolism , Thrombosis/chemically induced
6.
Blood ; 128(24): 2834-2845, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27694320

ABSTRACT

Activated factor XII (FXIIa) has plasminogen activator capacity but its relative contribution to fibrinolysis is considered marginal compared with urokinase and tissue plasminogen activator. Polyphosphate (polyP) is released from activated platelets and mediates FXII activation. Here, we investigate the contribution of polyP to the plasminogen activator function of αFXIIa. We show that both polyP70, of the chain length found in platelets (60-100 mer), and platelet-derived polyP significantly augment the plasminogen activation capacity of αFXIIa. PolyP70 stimulated the autoactivation of FXII and subsequent plasminogen activation, indicating that once activated, αFXIIa remains bound to polyP70 Indeed, complex formation between polyP70 and αFXIIa provides protection against autodegradation. Plasminogen activation by ßFXIIa was minimal and not enhanced by polyP70, highlighting the importance of the anion binding site. PolyP70 did not modulate plasmin activity but stimulated activation of Glu and Lys forms of plasminogen by αFXIIa. Accordingly, polyP70 was found to bind to FXII, αFXIIa, and plasminogen, but not ßFXIIa. Fibrin and polyP70 acted synergistically to enhance αFXIIa-mediated plasminogen activation. The plasminogen activator activity of the αFXIIa-polyP70 complex was modulated by C1 inhibitor and histidine-rich glycoprotein, but not plasminogen activator inhibitors 1 and 2. Platelet polyP and FXII were found to colocalize on the activated platelet membrane in a fibrin-dependent manner and decorated fibrin strands extending from platelet aggregates. We show that in the presence of platelet polyP and the downstream substrate fibrin, αFXIIa is a highly efficient and favorable plasminogen activator. Our data are the first to document a profibrinolytic function of platelet polyP.


Subject(s)
Blood Platelets/metabolism , Factor XII/metabolism , Fibrin/metabolism , Plasminogen/metabolism , Polyphosphates/metabolism , Blood Platelets/drug effects , Complement C1 Inhibitor Protein/pharmacology , Glutamic Acid/pharmacology , HeLa Cells , Humans , Lysine/pharmacology , Proteins/pharmacology
7.
J Vasc Surg ; 68(6S): 209S-221S.e2, 2018 12.
Article in English | MEDLINE | ID: mdl-29395422

ABSTRACT

OBJECTIVE: Ischemia-reperfusion (I/R) injury is a major clinical problem linked to vascular surgery. Currently, no drugs to prevent or to treat I/R injury are approved for clinical use. C1 inhibitor (C1 INH) is known to reduce activation of the plasma cascade systems that are involved in the pathophysiologic process of I/R injury. The aim of this study was therefore to investigate the effect of C1 INH on complement deposition and endothelial cell activation in a rat model of hind limb I/R injury. METHODS: Male Wistar rats (wild type, bred at the central animal facility, University of Bern), weighing 250 to 320 g, were used. The rats underwent 2-hour ischemia and 24-hour reperfusion by unilateral clamping of the femoral artery and additional use of a tourniquet. Five groups were divided according to intravenous treatment 5 minutes before ischemia: 50 IU/kg C1 INH (n = 5); 100 IU/kg C1 INH (n = 7); vehicle control (n = 5); nontreated control (n = 7); and normal, healthy control without intervention (n = 4). At the end, muscle edema, tissue viability, and histologic features were assessed. Deposition of immunoglobulin M, C1r, C4d, and fibrin and expression of plasminogen activator inhibitor 1, heparan sulfate (HS), E-selectin, and vascular cell adhesion molecule 1 were evaluated by fluorescence staining. In addition, high-mobility group box 1 protein was measured in plasma. RESULTS: Edema formation was reduced by C1 INH at two dosages, mirrored by improved histologic injury scores and preserved muscle viability. Deposition of immunoglobulin M, C4d, and fibrin was significantly decreased by 100 IU/kg C1 INH compared with nontreated controls. Pretreatment with 100 IU/kg C1 INH also significantly reduced HS shedding and expression of plasminogen activator inhibitor 1 as well as plasma levels of high-mobility group box 1 protein. CONCLUSIONS: Pretreatment with both 50 and 100 IU/kg C1 INH attenuated reperfusion injury of rat hind limbs. Pretreatment with 100 IU/kg also preserved the endothelial HS layer as well as the natural, profibrinolytic phenotype of the endothelium. Prevention of endothelial cell activation by C1 INH may therefore be a promising strategy to prevent I/R injury in the clinical setting of peripheral vascular diseases and elective surgery on extremities.


Subject(s)
Complement Activation/drug effects , Complement C1 Inhibitor Protein/pharmacology , Complement Inactivating Agents/pharmacology , Endothelial Cells/drug effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Reperfusion Injury/prevention & control , Animals , Complement C1r/metabolism , Complement C4b/metabolism , Disease Models, Animal , E-Selectin/metabolism , Edema/immunology , Edema/metabolism , Edema/pathology , Edema/prevention & control , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrin/metabolism , HMGB1 Protein/metabolism , Heparitin Sulfate/metabolism , Hindlimb , Immunoglobulin M/metabolism , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Peptide Fragments/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Rats, Wistar , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tissue Survival/drug effects , Vascular Cell Adhesion Molecule-1/metabolism
8.
J Thromb Thrombolysis ; 46(2): 219-226, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29860607

ABSTRACT

The crosstalk between immune and coagulation systems plays pivotal roles in host defense, which may involve monocyte-derived dendritic cells (moDCs). Our objectives were to elucidate the role of moDCs in coagulation under inflammatory conditions and the involvement of the complement system. We assessed the effects of lipopolysaccharide (LPS)-stimulated moDCs on coagulation using whole blood thromboelastometry in the presence of complement inhibitors. The sum of clotting time and clot formation time (CT plus CFT) in whole blood thromboelastometry was significantly more reduced in the presence of moDCs than in the absence of monocytes or moDCs and in the presence of monocytes, indicating a more potent coagulability of moDCs. The mRNA expression of coagulation-related proteins in moDCs was analyzed by quantitative PCR, which showed an increase only in the mRNA levels of tissue factor (TF). TF protein expression was assessed by western blot analysis and an activity assay, revealing higher TF expression in moDCs than that in monocytes. The in vitro moDC-associated hypercoagulable state was suppressed by a TF-neutralizing antibody, whereas LPS enhanced the in vitro hypercoagulation further. C1 inhibitor suppressed the in vitro LPS-enhanced whole blood hypercoagulability in the presence of moDCs and the increased TF expression in moDCs. These results suggest a significant role of moDCs and the complement system through TF expression in a hypercoagulable state under inflammatory conditions and demonstrate the suppressive effects of C1 inhibitor on moDC-associated hypercoagulation.


Subject(s)
Dendritic Cells/metabolism , Thrombophilia/etiology , Thromboplastin/metabolism , Blood Coagulation , Complement C1 Inhibitor Protein/pharmacology , Complement System Proteins , Dendritic Cells/drug effects , Humans , Inflammation , Lipopolysaccharides , Monocytes , RNA, Messenger/blood , Thrombelastography , Thrombophilia/genetics , Thromboplastin/genetics
9.
Am J Respir Crit Care Med ; 196(2): 186-199, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28005404

ABSTRACT

RATIONALE: Acute respiratory distress syndrome is characterized by alveolar epithelial cell injury, edema formation, and intraalveolar contact phase activation. OBJECTIVES: To explore whether C1 esterase inhibitor (C1INH), an endogenous inhibitor of the contact phase, may protect from lung injury in vivo and to decipher the possible underlying mechanisms mediating protection. METHODS: The ability of C1INH to control the inflammatory processes was studied in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Here, we demonstrate that application of C1INH alleviates bleomycin-induced lung injury via direct interaction with extracellular histones. In vitro, C1INH was found to bind all histone types. Interaction with histones was independent of its protease inhibitory activity, as demonstrated by the use of reactive-center-cleaved C1INH, but dependent on its glycosylation status. C1INH sialylated-N- and -O-glycans were not only essential for its interaction with histones but also to protect against histone-induced cell death. In vivo, histone-C1INH complexes were detected in bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome and multiple models of lung injury. Furthermore, reactive-center-cleaved C1INH attenuated pulmonary damage evoked by intravenous histone instillation. CONCLUSIONS: Collectively, C1INH administration provides a new therapeutic option for disorders associated with histone release.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Histones/metabolism , Lung Injury/prevention & control , Respiratory Distress Syndrome/physiopathology , Animals , Bronchoalveolar Lavage Fluid , Complement C1 Inhibitor Protein/metabolism , Disease Models, Animal , Humans , Lung/metabolism , Lung/physiopathology , Lung Injury/physiopathology , Mice , Mice, Inbred C57BL
10.
J Obstet Gynaecol Res ; 44(10): 1995-1998, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29974607

ABSTRACT

Amniotic fluid embolism (AFE) causes consumption coagulopathy, which requires a massive transfusion to save the mother's life. The preparation of such a massive transfusion is too time-consuming in extremely emergent clinical settings and occasionally leads to devastating side effects such as transfusion-associated acute lung injury. C1 esterase inhibitor (C1INH) is a protein with the ability to inhibit complement, coagulation and kinin pathways. The C1INH concentration in AFE patients is low, and it has been speculated that the administration of C1INH concentrate could have a striking and beneficial effect on AFE patients in critical condition by ameliorating their perturbed coagulation system. We report the case of a 32-year-old Japanese AFE patient in whom deteriorated vital signs and coagulopathy recovered within minutes after an injection of C1INH concentrate. C1INH concentrate can quickly revive the deteriorated vital signs and the atonic uterus that stem from AFE and may reduce the total amount of transfusion.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Embolism, Amniotic Fluid/drug therapy , Hematologic Agents/pharmacology , Adult , Cesarean Section , Complement C1 Inhibitor Protein/administration & dosage , Female , Hematologic Agents/administration & dosage , Humans , Pregnancy
11.
Am J Transplant ; 16(12): 3468-3478, 2016 12.
Article in English | MEDLINE | ID: mdl-27184779

ABSTRACT

Antibody-mediated rejection (AMR) is typically treated with plasmapheresis (PP) and intravenous immunoglobulin (standard of care; SOC); however, there is an unmet need for more effective therapy. We report a phase 2b, multicenter double-blind randomized placebo-controlled pilot study to evaluate the use of human plasma-derived C1 esterase inhibitor (C1 INH) as add-on therapy to SOC for AMR. Eighteen patients received 20 000 units of C1 INH or placebo (C1 INH n = 9, placebo n = 9) in divided doses every other day for 2 weeks. No discontinuations, graft losses, deaths, or study drug-related serious adverse events occurred. While the study's primary end point, a difference between groups in day 20 pathology or graft survival, was not achieved, the C1 INH group demonstrated a trend toward sustained improvement in renal function. Six-month biopsies performed in 14 subjects (C1 INH = 7, placebo = 7) showed no transplant glomerulopathy (TG) (PTC+cg≥1b) in the C1 INH group, whereas 3 of 7 placebo subjects had TG. Endogenous C1 INH measured before and after PP demonstrated decreased functional C1 INH serum concentration by 43.3% (p < 0.05) for both cohorts (C1 INH and placebo) associated with PP, although exogenous C1 INH-treated patients achieved supraphysiological levels throughout. This new finding suggests that C1 INH replacement may be useful in the treatment of AMR.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Complement Inactivating Agents/pharmacology , Graft Rejection/drug therapy , Isoantibodies/adverse effects , Kidney Transplantation/adverse effects , Double-Blind Method , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival , Humans , Immunoglobulins, Intravenous/administration & dosage , Kidney Failure, Chronic/surgery , Kidney Function Tests , Male , Middle Aged , Pilot Projects , Plasmapheresis , Prognosis , Risk Factors
12.
Clin Exp Immunol ; 184(3): 378-88, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26874675

ABSTRACT

Complement activation is of major importance in numerous pathological conditions. Therefore, targeted complement inhibition is a promising therapeutic strategy. C1-esterase inhibitor (C1-INH) controls activation of the classical pathway (CP) and the lectin pathway (LP). However, conflicting data exist on inhibition of the alternative pathway (AP) by C1-INH. The inhibitory capacity of C1-INH for the CP is potentiated by heparin and other glycosaminoglycans, but no data exist for the LP and AP. The current study investigates the effects of C1-INH in the presence or absence of different clinically used heparinoids on the CP, LP and AP. Furthermore, the combined effects of heparinoids and C1-INH on coagulation were investigated. C1-INH, heparinoids or combinations were analysed in a dose-dependent fashion in the presence of pooled serum. Functional complement activities were measured simultaneously using the Wielisa(®) -kit. The activated partial thrombin time was determined using an automated coagulation analyser. The results showed that all three complement pathways were inhibited significantly by C1-INH or heparinoids. Next to their individual effects on complement activation, heparinoids also enhanced the inhibitory capacity of C1-INH significantly on the CP and LP. For the AP, significant potentiation of C1-INH by heparinoids was found; however, this was restricted to certain concentration ranges. At low concentrations the effect on blood coagulation by combining heparinoids with C1-INH was minimal. In conclusion, our study shows significant potentiating effects of heparinoids on the inhibition of all complement pathways by C1-INH. Therefore, their combined use is a promising and a potentially cost-effective treatment option for complement-mediated diseases.


Subject(s)
Complement Activation/drug effects , Complement C1 Inhibitor Protein/pharmacology , Heparinoids/pharmacology , Blood Coagulation/drug effects , Complement Pathway, Alternative/drug effects , Complement Pathway, Classical/drug effects , Complement Pathway, Mannose-Binding Lectin/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Humans , Partial Thromboplastin Time
13.
Allergy ; 70(3): 285-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25477154

ABSTRACT

BACKGROUND: Little is known about factors which trigger and/or contribute to hereditary angioedema or ACE-inhibitor-mediated angioedema including variations in bradykinin type 2 receptor (B2R) expression and activity. METHODS: Protein and mRNA expression of B2R and the increase of intracellular calcium (iCa) in response to bradykinin were monitored in porcine and murine endothelial cells in response to NO donors or bradykinin. B2R protein expression was evaluated in skin, heart, and lung of (i) mice with endothelial-specific overexpression of eNOS (eNOS(tg) ), (ii) in eNOS(-/-) mice and (iii) in C57BL/6 mice treated with the NO donor pentaerythritol tetranitrate (PETN), the NOS inhibitor l-nitroarginine (L-NA), plasma pool C1-INH, and the B2R antagonist icatibant. Aortic reactivity to bradykinin was investigated including eNOS(-/-) mice. RESULTS: B2R protein and mRNA expression remained unchanged in cells subjected to L-NA, NO donors, and bradykinin in a time- and concentration-dependent manner. Likewise, increases of iCa in murine brain endothelial cells remained unchanged. B2R protein levels were similar in eNOS(tg) and eNOS(-/-) as compared to transgene-negative littermates. Likewise, treatment of C57BL/6 mice with PETN, L-NA, C1-INH or icatibant did not change B2R protein expression. In aortic rings of C57BL/6 mice, bradykinin induced B2R-dependent constrictions which were attenuated by endothelial NO and abolished by diclofenac indicating the functional importance of B2R-induced activation of endothelial NO synthase and cyclooxygenase. CONCLUSION: These data suggest that alterations of B2R protein expression induced by NO, bradykinin, C1-INH, or icatibant unlikely contribute to bradykinin-induced angioedema. This finding does not rule out a role for NO in bradykinin-induced extravasation and/or angioedema.


Subject(s)
Bradykinin B2 Receptor Antagonists/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Complement C1 Inhibitor Protein/pharmacology , Nitric Oxide/metabolism , Receptor, Bradykinin B2/metabolism , Animals , Bradykinin/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Mice, Knockout , Mice, Transgenic , Receptor, Bradykinin B2/genetics , Swine
14.
Cytokine ; 70(2): 173-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25148723

ABSTRACT

In order to examine the immunomodulatory effects of antithrombin III (AT-III) and C1 esterase inhibitor (C1-INH) in human monocytes, we investigated the intracellular expression of interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α in an ex-vivo laboratory study in a whole blood setting. Heparinized whole blood samples from 23 healthy male and female volunteers (mean age: 27±7years) were pre-incubated with clinically relevant concentrations of AT-III (n=11) and C1-INH (n=12), then stimulated with 0.2 ng/mL lipopolysaccharide (LPS) for 3h. After phenotyping CD14⁺ monocytes, intracellular expression of IL-6, IL-8, and TNF-α was assessed using flow cytometry. In addition, 12 whole blood samples (AT-III and C1-INH, n=6 each) were examined using hirudin for anticoagulation; all samples were processed in the same way. To exclude cytotoxicity effects, 7-amino-actinomycin D and Nonidet P40 staining were used to investigate probes. This study is the first to demonstrate the influence of C1-INH and AT-III on the monocytic inflammatory response in a whole blood setting, which mimics the optimal physiological setting. Cells treated with AT-III exhibited significant downregulation of the proportion of gated CD14⁺ monocytes for IL-6 and IL-8, in a dose-dependent manner; downregulation for TNF-α did not reach statistical significance. There were no significant effects on mean fluorescence intensity (MFI). In contrast, C1-INH did not significantly reduce the proportion of gated CD14⁺ monocytes or the MFI regarding IL-6, TNF-α, and IL-8. When using hirudin for anticoagulation, no difference in the anti-inflammatory properties of AT-III and C1-INH in monocytes occurs. Taken together, in contrast to TNF-α, IL-6 and IL-8 were significantly downregulated in monocytes in an ex-vivo setting of human whole blood when treated with AT-III. This finding implicates monocytes as an important point of action regarding the anti-inflammatory properties of AT-III in sepsis. C1-INH was unable to attenuate the monocytic response, which supports the hypothesis that other cellular components in whole blood (e.g., neutrophils) might be responsible for the known effects of C1-INH in inflammation.


Subject(s)
Antithrombin III/pharmacology , Complement C1 Inhibitor Protein/pharmacology , Inflammation/blood , Inflammation/pathology , Lipopolysaccharides/pharmacology , Monocytes/pathology , Adult , Anticoagulants/pharmacology , Cell Death/drug effects , Female , Hirudins/pharmacology , Humans , Male , Monocytes/drug effects , Young Adult
15.
J Surg Res ; 187(2): 660-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24433870

ABSTRACT

Liver ischemia-reperfusion injury (IRI) is a well-known cause of morbidity and mortality after liver transplantation (LT). Activation of the complement system contributes to the pathogenesis of IRI. Effective treatment strategies aimed at reducing hepatic IRI and accelerating liver regeneration could offer major benefits in LT. Herein, we investigated the effect of C1-esterase inhibitor (human) [C1-INH] on IRI and liver regeneration. Mice were subjected to 60-min partial IRI, with or without 70% partial hepatectomy, or CCl4-induced acute liver failure. Before liver injury, the animals were pretreated with intravenous C1-INH or normal saline. Liver IRI was evaluated using serum levels of alanine aminotransferase, serum interleukin-6, and histopathology. Liver samples were stained for specific markers of regeneration (5-bromo-2'-deoxyuridine [BrdU] staining and proliferating cell nuclear antigen [PCNA]). Histology, serum interleukin-6, and alanine aminotransferase release revealed that C1-INH treatment attenuated liver injury compared with controls. Improved animal survival and increased number of BrdU- and PCNA-positive cells were observed in C1-INH-treated animals which underwent IRI + partial hepatectomy or CCl4 injection compared with control group. These data indicate that complement plays a key role in IRI and liver regeneration. C1-INH represents a potential therapeutic strategy to reduce IRI and promote regeneration in LT.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Liver Failure, Acute/drug therapy , Liver Failure, Acute/etiology , Liver Regeneration/drug effects , Liver Transplantation/adverse effects , Reperfusion Injury/drug therapy , Animals , Complement C1 Inhibitor Protein/metabolism , Complement C3/genetics , Hepatectomy/methods , Humans , Injections, Intravenous , Liver/drug effects , Liver/physiology , Liver/surgery , Liver Failure, Acute/mortality , Liver Regeneration/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/mortality , Reperfusion Injury/physiopathology , Toll-Like Receptor 4/genetics
16.
Adv Exp Med Biol ; 735: 23-40, 2013.
Article in English | MEDLINE | ID: mdl-23402017

ABSTRACT

Proteases play important roles in human physiology and pathology. The complement system is a proteolytic cascade, where serine proteases activate each other by limited proteolysis in a strictly ordered manner. Serine proteases are essential in both the initiation and the amplification of the cascade. Since uncontrolled complement activation contributes to the development of serious disease conditions, inhibition of the complement serine proteases could be an attractive therapeutic approach. In this chapter, we give a brief overview of the major types of natural serine protease inhibitors and their role in controlling the complement cascade. A special emphasis is laid on C1-inhibitor, a natural complement protease inhibitor, which is approved for clinical use in hereditary angioedema (HAE). We also examine the potential of developing artificial complement protease inhibitors. Synthetic small-molecule drugs can be very efficient serine protease inhibitors, but they usually lack sufficient specificity. A promising approach to yield more specific compounds is the alteration of natural protease inhibitors through engineering or directed evolution resulting in new variants with fine-tuned specificity and enhanced affinity.


Subject(s)
Complement System Proteins/physiology , Serine Proteinase Inhibitors/pharmacology , Animals , Complement C1 Inhibitor Protein/pharmacology , Humans , Protein Engineering , Serine Proteases/physiology , Serine Proteinase Inhibitors/therapeutic use , Small Molecule Libraries
17.
J Clin Immunol ; 32(6): 1204-12, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22797816

ABSTRACT

Hereditary angioedema (HAE) is a rare disorder caused by a deficiency of C1 esterase inhibitor, characterized by recurrent, highly variable attacks of subcutaneous or submucosal edema that may affect multiple body sites. Clinical studies of acute HAE therapies have required the use of assessment tools to evaluate both pretreatment attack severity (baseline severity) and changes in symptom severity following treatment (treatment response). This article reviews the range of assessment tools used for efficacy determination of acute HAE therapies, based on a review of relevant clinical studies. Because the goal is relief of symptoms (rather than cure), patient-reported outcomes (PROs) form the basis of these tools. Tools used to evaluate baseline severity typically employ location-specific assessment of symptom severity, using either categorical descriptions (which may be converted into numerical variables) or a visual analog scale (VAS). Some studies define the initial or most symptomatic site as an "index" site for purposes of efficacy determination, while others (such as the Mean Symptom Complex Severity score used in clinical studies of ecallantide) use a composite score that reflects all sites. Assessment of treatment response typically employs the same tool(s) to evaluate baseline severity, and may be either time-based (e.g., time to achievement of minimal or no symptoms) or symptom-based (e.g., degree of symptom relief at predetermined time points). Although it is unlikely that therapies will be compared using identical assessment tools, prospective or retrospective validation ensures the adequacy and relevance of such tools, which should be taken into consideration when therapies are compared.


Subject(s)
Complement C1 Inhibitor Protein/therapeutic use , Hereditary Angioedema Types I and II/psychology , Outcome Assessment, Health Care , Peptides/therapeutic use , Research Design , Complement C1 Inhibitor Protein/pharmacology , Hereditary Angioedema Types I and II/drug therapy , Hereditary Angioedema Types I and II/physiopathology , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Pain Measurement , Peptides/pharmacology , Prospective Studies , Randomized Controlled Trials as Topic , Retrospective Studies , Severity of Illness Index , Time Factors , Treatment Outcome
18.
Article in English | MEDLINE | ID: mdl-32614753

ABSTRACT

BACKGROUND: Burns induce a boost in local and systemic complement levels as well as immune cell infiltration in the burn wound, which may negatively affect wound healing. OBJECTIVE: In this study, the effects of long-term treatment with complement inhibitor C1 esterase inhibitor (C1inh) on post-burn inflammation and wound healing parameters were analyzed in time up to 60 days post-burn. METHODS: Burned pigs were treated either with or without C1inh up to 15 days post-burn. Burn wound biopsies and blood were collected at different time points up to 60 days post-burn. Thereafter, complement in blood as well as complement and immune cells in the wound, capillary leakage, necrosis, reepithelialization and wound contraction were quantified. RESULTS: No significant differences in complement C3 blood levels were observed at any time point between C1inh-treated and control pigs. In the wound, complement C4 levels were significantly lower in the C1inh group than in controls at day 3-6 and 21-30 post-burn. Similarly, C3 levels, neutrophil and macrophage infiltration in the wound were, although not statistically significant, reduced in C1inh-treated pigs at day 9-14 post-burn. No differences in lymphocyte infiltration in the wound were found between C1inh and control pigs. C1inh-treated pigs also showed reduced capillary leakage. Despite these effects, no significant differences in the long-term wound healing parameters necrosis, reepithelialization and wound contraction were observed between C1inh and control pigs. CONCLUSION: In pigs, 15 days of C1inh treatment after burn, leads to a reduction in local inflammation and capillary leakage in the burn wound without affecting long-term wound healing parameters.


Subject(s)
Burns/drug therapy , Complement C1 Inhibitor Protein/pharmacology , Inflammation/drug therapy , Wound Healing/drug effects , Animals , Burns/physiopathology , Disease Models, Animal , Female , Inflammation/physiopathology , Random Allocation , Swine
19.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G878-83, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20338925

ABSTRACT

The complement and contact systems may be involved in the pathophysiological process of inflammatory bowel disease (IBD). C1 inhibitor (C1INH) is the most important inhibitor of both the complement and contact systems. We evaluated the role of these systems and the effect of both active and inactive forms of C1INH (iC1INH) in dextran sulfate sodium (DSS)-induced colitis mouse model. Three percent DSS was used in drinking water to induce colitis in complement C3-deficient (C3(-/-)) mice, bradykinin type 2 receptor deficient (Bk(2)R(-/-)) mice, and C57BL/6 mice. After ten days DSS exposure, C3(-/-) mice exhibited markedly less weight loss than wild-type (WT) mice (12 +/- 3.3% vs. 30 +/- 1.2%, P < 0.05) and developed a milder disease-activity index (DAI), histological score, colon shortening, and myeloperoxidase (MPO) elevation (P < 0.05, respectively). The Bk(2)R(-/-) mice were not protected from the disease. Seven-day treatment with either native C1INH or iC1INH reduced the severity of the disease in WT mice, as indicated by decreased weight loss (15 +/- 1.8%, 14 +/- 2.1% vs. 30 +/- 1.2%, P < 0.05, respectively), DAI, intestinal tissue damage, and MPO elevation compared with untreated WT DSS control mice (P < 0.05, respectively). These findings suggest that complement plays a role in the development of DSS-induced colitis and that blockade of the complement system might be useful for the acute phase of IBD treatment. C1INH, however, leads to an amelioration of DSS-induced colitis via a mechanism that does not involve the inhibition of complement or contact system activation but does result in significant suppression of leukocyte infiltration.


Subject(s)
Colitis/chemically induced , Colitis/drug therapy , Complement C1 Inhibitor Protein/pharmacology , Dextran Sulfate/toxicity , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Animals , Colitis/metabolism , Colon/enzymology , Colon/pathology , Complement C3/deficiency , Complement C3/genetics , Complement C3/metabolism , Disease Models, Animal , Inflammatory Bowel Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Time Factors
20.
Ann Neurol ; 66(3): 332-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19798727

ABSTRACT

OBJECTIVE: C1 inhibitor (C1-INH) is an endogenous inhibitor of complement and kinin systems. We have explored the efficacy and the therapeutic window of the recently available human recombinant (rh) C1-INH on ischemic brain injury and investigated its mechanism of action in comparison with that of plasma-derived (pd) C1-INH. METHODS: rhC1-INH was administered intravenously to C57Bl/6 mice undergoing transient or permanent ischemia, and its protective effects were evaluated by measuring infarct volume and neurodegeneration. The binding profiles of rhC1-INH and pdC1-INH were assessed in vitro using surface plasmon resonance. Their localization in the ischemic brain tissue was determined by immunohistochemistry and confocal analysis. The functional consequences of rhC1-INH and pdC1-INH administration on complement activation were analyzed by enzyme-linked immunosorbent assay on plasma samples. RESULTS: rhC1-INH markedly reduced cerebral damage when administered up to 18 hours after transient ischemia and up to 6 hours after permanent ischemia, thus showing a surprisingly wide therapeutic window. In vitro rhC1-INH bound mannose-binding lectin (MBL), a key protein in the lectin complement pathway, with high affinity, whereas pdC1-INH, which has a different glycosylation pattern, did not. In the ischemic brain, rhC1-INH was confined to cerebral vessels, where it colocalized with MBL, whereas pdC1-INH diffused into the brain parenchyma. In addition, rhC1-INH was more active than pdC1-INH in inhibiting MBL-induced complement activation. INTERPRETATION: rhC1-INH showed a surprisingly wider time window of efficacy compared with the corresponding plasmatic protein. We propose that the superiority of rhC1-INH is due to its selective binding to MBL, which emerged as a novel target for stroke treatment.


Subject(s)
Brain Ischemia/drug therapy , Complement C1 Inhibitor Protein/therapeutic use , Ischemic Attack, Transient/drug therapy , Animals , Brain Ischemia/metabolism , Complement Activation , Complement C1 Inhibitor Protein/pharmacology , Complement Pathway, Alternative , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mannose-Binding Lectin/pharmacology , Mice , Mice, Inbred C57BL , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL