Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
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
2.
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
3.
J Neuroinflammation ; 14(1): 39, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28219400

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a devastating neurological condition and a frequent cause of permanent disability. Posttraumatic inflammation and brain edema formation, two pathological key events contributing to secondary brain injury, are mediated by the contact-kinin system. Activation of this pathway in the plasma is triggered by activated factor XII. Hence, we set out to study in detail the influence of activated factor XII on the abovementioned pathophysiological features of TBI. METHODS: Using a cortical cryogenic lesion model in mice, we investigated the impact of genetic deficiency of factor XII and inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused Infestin-4 on the release of bradykinin, the brain lesion size, and contact-kinin system-dependent pathological events. We determined protein levels of bradykinin, intracellular adhesion molecule-1, CC-chemokine ligand 2, and interleukin-1ß by enzyme-linked immunosorbent assays and mRNA levels of genes related to inflammation by quantitative real-time PCR. Brain lesion size was determined by tetrazolium chloride staining. Furthermore, protein levels of the tight junction protein occludin, integrity of the blood-brain barrier, and brain water content were assessed by Western blot analysis, extravasated Evans Blue dye, and the wet weight-dry weight method, respectively. Infiltration of neutrophils and microglia/activated macrophages into the injured brain lesions was quantified by immunohistological stainings. RESULTS: We show that both genetic deficiency of factor XII and inhibition of activated factor XII in mice diminish brain injury-induced bradykinin release by the contact-kinin system and minimize brain lesion size, blood-brain barrier leakage, brain edema formation, and inflammation in our brain injury model. CONCLUSIONS: Stimulation of bradykinin release by activated factor XII probably plays a prominent role in expanding secondary brain damage by promoting brain edema formation and inflammation. Pharmacological blocking of activated factor XII could be a useful therapeutic principle in the treatment of TBI-associated pathologic processes by alleviating posttraumatic inflammation and brain edema formation.


Subject(s)
Brain Edema/metabolism , Brain Edema/prevention & control , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/prevention & control , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Animals , Bradykinin/metabolism , Brain Injuries/metabolism , Brain Injuries/prevention & control , Factor XIIa/genetics , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Ann Neurol ; 79(6): 970-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27043916

ABSTRACT

OBJECTIVE: Traumatic brain injury is a major global public health problem for which specific therapeutic interventions are lacking. There is, therefore, a pressing need to identify innovative pathomechanism-based effective therapies for this condition. Thrombus formation in the cerebral microcirculation has been proposed to contribute to secondary brain damage by causing pericontusional ischemia, but previous studies have failed to harness this finding for therapeutic use. The aim of this study was to obtain preclinical evidence supporting the hypothesis that targeting factor XII prevents thrombus formation and has a beneficial effect on outcome after traumatic brain injury. METHODS: We investigated the impact of genetic deficiency of factor XII and acute inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused infestin-4 (rHA-Infestin-4) on trauma-induced microvascular thrombus formation and the subsequent outcome in 2 mouse models of traumatic brain injury. RESULTS: Our study showed that both genetic deficiency of factor XII and an inhibition of activated factor XII in mice minimize trauma-induced microvascular thrombus formation and improve outcome, as reflected by better motor function, reduced brain lesion volume, and diminished neurodegeneration. Administration of human factor XII in factor XII-deficient mice fully restored injury-induced microvascular thrombus formation and brain damage. INTERPRETATION: The robust protective effect of rHA-Infestin-4 points to a novel treatment option that can decrease ischemic injury after traumatic brain injury without increasing bleeding tendencies. Ann Neurol 2016;79:970-982.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Factor XII/therapeutic use , Factor XIIa/antagonists & inhibitors , Insect Proteins/therapeutic use , Intracranial Thrombosis/drug therapy , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/therapeutic use , Adult , Aged , Animals , Brain Injuries, Traumatic/physiopathology , Case-Control Studies , Disease Models, Animal , Factor XII/genetics , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Middle Aged , Neuroimaging , Platelet Aggregation/physiology , Serum Albumin, Human , Young Adult
5.
Br J Haematol ; 173(5): 769-78, 2016 06.
Article in English | MEDLINE | ID: mdl-27018425

ABSTRACT

Haemostasis including blood coagulation is initiated upon vessel wall injury and indispensable to limit excessive blood loss. However, unregulated pathological coagulation may lead to vessel occlusion, causing thrombotic disorders, most notably myocardial infarction and stroke. Furthermore, blood exposure to foreign surfaces activates the intrinsic pathway of coagulation. Hence, various clinical scenarios, such as extracorporeal membrane oxygenation, require robust anticoagulation consequently leading to an increased bleeding risk. This study aimed to further assess the antithrombotic efficacy of the activated factor XII (FXIIa) inhibitor, rHA-Infestin-4, in several thrombosis models. In mice, rHA-Infestin-4 decreased occlusion rates in the mechanically-induced arterial (Folt's) and the FeCl3 -induced venous thrombosis model. rHA-Infestin-4 also protected from FeCl3 -induced arterial thrombosis and from stasis-prompted venous thrombosis in rabbits. Furthermore, rHA-Infestin-4 prevented occlusion in the arterio-venous shunt model in mice and rabbits where thrombosis was induced via a foreign surface. In contrast to heparin, the haemostatic capacity in rabbits was unaffected by rHA-Infestin-4. Using rodent and non-rodent species, our data demonstrate that the FXIIa inhibitor rHA-Infestin-4 decreased arterial, venous and foreign surface-induced thrombosis without affecting physiological haemostasis. Hence, we provide further evidence that targeting FXIIa represents a potent yet safe antithrombotic treatment approach, especially in foreign surface-triggered thrombosis.


Subject(s)
Factor XIIa/antagonists & inhibitors , Insect Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Serum Albumin/pharmacology , Thrombosis/drug therapy , Animals , Arterial Occlusive Diseases/drug therapy , Arterial Occlusive Diseases/etiology , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Hemostasis/drug effects , Insect Proteins/therapeutic use , Kinetics , Mice , Rabbits , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/therapeutic use , Serum Albumin, Human , Thrombosis/etiology , Treatment Outcome , Venous Thrombosis/drug therapy , Venous Thrombosis/etiology
6.
Haematologica ; 101(4): 427-36, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26721892

ABSTRACT

Coated platelets, formed by collagen and thrombin activation, have been characterized in different ways: i) by the formation of a protein coat of α-granular proteins; ii) by exposure of procoagulant phosphatidylserine; or iii) by high fibrinogen binding. Yet, their functional role has remained unclear. Here we used a novel transglutaminase probe, Rhod-A14, to identify a subpopulation of platelets with a cross-linked protein coat, and compared this with other platelet subpopulations using a panel of functional assays. Platelet stimulation with convulxin/thrombin resulted in initial integrin α(IIb)ß3 activation, the appearance of a platelet population with high fibrinogen binding, (independently of active integrins, but dependent on the presence of thrombin) followed by phosphatidylserine exposure and binding of coagulation factors Va and Xa. A subpopulation of phosphatidylserine-exposing platelets bound Rhod-A14 both in suspension and in thrombi generated on a collagen surface. In suspension, high fibrinogen and Rhod-A14 binding were antagonized by combined inhibition of transglutaminase activity and integrin α(IIb)ß3 Markedly, in thrombi from mice deficient in transglutaminase factor XIII, platelet-driven fibrin formation and Rhod-A14 binding were abolished by blockage of integrin α(IIb)ß3. Vice versa, star-like fibrin formation from platelets of a patient with deficiency in α(IIb)ß3(Glanzmann thrombasthenia) was abolished upon blockage of transglutaminase activity. We conclude that coated platelets, with initial α(IIb)ß3 activation and high fibrinogen binding, form a subpopulation of phosphatidylserine-exposing platelets, and function in platelet-dependent star-like fibrin fiber formation via transglutaminase factor XIII and integrin α(IIb)ß3.


Subject(s)
Blood Platelets/metabolism , Factor XIII/metabolism , Fibrin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombasthenia/blood , Animals , Blood Coagulation , Blood Platelets/drug effects , Blood Platelets/pathology , Crotalid Venoms/pharmacology , Factor Va/chemistry , Factor Va/metabolism , Factor XIII/chemistry , Factor Xa/chemistry , Factor Xa/metabolism , Fibrin/chemistry , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Lectins, C-Type , Mice , Mice, Knockout , Molecular Probes/chemistry , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Primary Cell Culture , Protein Binding , Thrombasthenia/pathology , Thrombin/pharmacology
7.
Circulation ; 127(20): 2038-46, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23616627

ABSTRACT

BACKGROUND: Exaggerated and prolonged inflammation after myocardial infarction (MI) accelerates left ventricular remodeling. Inflammatory pathways may present a therapeutic target to prevent post-MI heart failure. However, the appropriate magnitude and timing of interventions are largely unknown, in part because noninvasive monitoring tools are lacking. Here, we used nanoparticle-facilitated silencing of CCR2, the chemokine receptor that governs inflammatory Ly-6C(high) monocyte subset traffic, to reduce infarct inflammation in apolipoprotein E-deficient (apoE(-/-)) mice after MI. We used dual-target positron emission tomography/magnetic resonance imaging of transglutaminase factor XIII (FXIII) and myeloperoxidase (MPO) activity to monitor how monocyte subset-targeted RNAi altered infarct inflammation and healing. METHODS AND RESULTS: Flow cytometry, gene expression analysis, and histology revealed reduced monocyte numbers and enhanced resolution of inflammation in infarcted hearts of apoE(-/-) mice that were treated with nanoparticle-encapsulated siRNA. To follow extracellular matrix cross-linking noninvasively, we developed a fluorine-18-labeled positron emission tomography agent ((18)F-FXIII). Recruitment of MPO-rich inflammatory leukocytes was imaged with a molecular magnetic resonance imaging sensor of MPO activity (MPO-Gd). Positron emission tomography/magnetic resonance imaging detected anti-inflammatory effects of intravenous nanoparticle-facilitated siRNA therapy (75% decrease of MPO-Gd signal; P<0.05), whereas (18)F-FXIII positron emission tomography reflected unimpeded matrix cross-linking in the infarct. Silencing of CCR2 during the first week after MI improved ejection fraction on day 21 after MI from 29% to 35% (P<0.05). CONCLUSION: CCR2-targeted RNAi reduced recruitment of Ly-6C(high) monocytes, attenuated infarct inflammation, and curbed post-MI left ventricular remodeling.


Subject(s)
Atherosclerosis/therapy , Gene Targeting/methods , Monocytes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , RNA Interference/physiology , Receptors, CCR2/genetics , Wound Healing/genetics , Amino Acid Sequence , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Female , Genetic Predisposition to Disease , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Monocytes/pathology , Myocardial Infarction/pathology , Random Allocation , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism
8.
Toxicol Appl Pharmacol ; 280(1): 70-7, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25102310

ABSTRACT

Fibrinogen, a soluble 340kDa plasma glycoprotein, is critical in achieving and maintaining hemostasis. Reduced fibrinogen levels are associated with an increased risk of bleeding and recent research has investigated the efficacy of fibrinogen concentrate for controlling perioperative bleeding. European guidelines on the management of perioperative bleeding recommend the use of fibrinogen concentrate if significant bleeding is accompanied by plasma fibrinogen levels less than 1.5-2.0g/l. Plasma-derived human fibrinogen concentrate has been available for therapeutic use since 1956. The overall aim of the comprehensive series of non-clinical investigations presented was to evaluate i) the pharmacodynamic and pharmacokinetic characteristics and ii) the safety and tolerability profile of human fibrinogen concentrate Haemocomplettan P® (RiaSTAP®). Pharmacodynamic characteristics were assessed in rabbits, pharmacokinetic parameters were determined in rabbits and rats and a safety pharmacology study was performed in beagle dogs. Additional toxicology tests included: single-dose toxicity tests in mice and rats; local tolerance tests in rabbits; and neoantigenicity tests in rabbits and guinea pigs following the introduction of pasteurization in the manufacturing process. Human fibrinogen concentrate was shown to be pharmacodynamically active in rabbits and dogs and well tolerated, with no adverse events and no influence on circulation, respiration or hematological parameters in rabbits, mice, rats and dogs. In these non-clinical investigations, human fibrinogen concentrate showed a good safety profile. This data adds to the safety information available to date, strengthening the current body of knowledge regarding this hemostatic agent.


Subject(s)
Fibrinogen/metabolism , Fibrinogen/toxicity , Animals , Body Weight/drug effects , Body Weight/physiology , Dogs , Dose-Response Relationship, Drug , Female , Fibrinogen/pharmacology , Guinea Pigs , Hemostasis/drug effects , Hemostasis/physiology , Humans , Male , Mice , Rabbits , Rats , Rats, Wistar , Species Specificity
10.
J Thromb Haemost ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39019440

ABSTRACT

BACKGROUND: The X-linked bleeding disorder Hemophilia B, caused by mutation(s) in the coagulation factor IX (FIX) gene, leads to partial or total loss of its function requiring lifelong FIX replacement therapy. Although new recombinant FIX (rIX) therapeutics like albumin-fusion proteins (rIX-FP) enable longer plasma half-life and thus less frequent administration, the complexity of intravenous (IV) injection remains. OBJECTIVES: The study aims to characterize rIX-FP variants with anticipated enhanced specific activity, which would leverage rIX-FP's superior pharmacokinetic (PK) profile with beneficial characteristics for subcutaneous (SC) administration. METHODS: Two rIX-FP variants, R338L ("Padua-variant") and R338L/E410K, were characterized in vitro. PK profiles of FIX antigen and activity levels were evaluated in FIX-deficient mice after IV and SC administration of these variants (dosing based on antigen levels). The efficacy of the most promising variant was tested after IV and SC administration (dosing based on activity) in a tail-clip bleeding model. A marketed wildtype (WT) rIX-FP product served as the comparator. RESULTS: Both rIX-FP variants showed a 4- to 5-fold increase in specific activity in vitro compared to rIX(WT)-FP, whilst FXIa-mediated activation was the fastest for rIX(WT)-FP and rIX(R338L)-FP. Compared to rIX(WT)-FP and rIX(R338L/E410K)-FP, rIX(R338L)-FP exhibited higher FIX activity exposure after IV and SC administration, and demonstrated comparable efficacy towards rIX(WT)-FP in reducing bleeding time and blood loss in FIX-deficient mice requiring ∼4 times lower protein amount. CONCLUSIONS: rIX(R338L)-FP was shown to be a promising candidate for SC administration, exhibiting increased specific activity combined with higher activity-based exposure, and indicating efficacy at lower protein dose.

11.
J Neurosci ; 31(10): 3926-34, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21389247

ABSTRACT

One major hallmark of Alzheimer's disease (AD) is the massive loss of synapses that occurs at an early clinical stage of the disease. In this study, we characterize alterations in spine density and the expression of synapse-associated immediate early gene Arc (activity-regulated cytoskeleton-associated protein) in the hippocampal CA1 regions of two different amyloid precursor protein (APP) transgenic mouse lines before plaque development and their connection to performance in hippocampus-dependent memory tests. The density of mushroom-type spines was reduced by 34% in the basal dendrites proximal to the soma of CA1 pyramidal neurons in 5.5-month-old Tg2576 mice, carrying the Swedish mutation, compared with wild-type littermates. A similar reduction of 42% was confirmed in the same region of 8-month-old APP/Lo mice, carrying the London mutation. In this strain, the reduction extended to the distal dendritic spines (28%), although no differences were found in apical dendrites in either transgenic mouse line. Both transgenic mice lines presented a significant increase in Arc protein expression in CA1 compared with controls, suggesting rather an overactivity and increased spine turnover that was supported by a significant decrease in number of somatostatin-immunopositive inhibitory interneurons in the stratum oriens of CA1. Behaviorally, the transgenic mice showed decrease freezing in the fear contextual conditioning test and impairment in spatial memory assessed by Morris water maze test. These data indicate that cognitive impairment in APP transgenic mice is correlated with impairment of synaptic connectivity in hippocampal CA1, probably attributable to loss of inhibitory interneurons and subsequent hyperactivity.


Subject(s)
Alzheimer Disease/metabolism , CA1 Region, Hippocampal/metabolism , Dendritic Spines/metabolism , Pyramidal Cells/metabolism , Alzheimer Disease/genetics , Analysis of Variance , Animals , Conditioning, Classical/physiology , Cytoskeletal Proteins/metabolism , Dendritic Spines/genetics , Disease Models, Animal , Fear/physiology , Freezing Reaction, Cataleptic/physiology , Immunohistochemistry , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Somatostatin/metabolism
12.
Stroke ; 43(9): 2457-67, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22744646

ABSTRACT

BACKGROUND AND PURPOSE: Inflammation and thrombosis are pathophysiological hallmarks of ischemic stroke still unamenable to therapeutic interventions. The contact-kinin system represents an interface between inflammatory and thrombotic circuits and is involved in stroke development. C1-inhibitor counteracts activation of the contact-kinin system at multiple levels. We investigated the therapeutic potential of C1-inhibitor in models of ischemic stroke. METHODS: Male and female C57Bl/6 mice and rats of different ages were subjected to middle cerebral artery occlusion and treated with C1-inhibitor after 1 hour or 6 hours. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, thrombus formation, and the local inflammatory response were determined poststroke. RESULTS: Treatment with 15.0 U C1-inhibitor, but not 7.5 U, 1 hour after stroke reduced infarct volumes by ≈60% and improved clinical scores in mice of either sex on day 1. This protective effect was preserved at later stages of infarction as well as in elderly mice and in another species, ie, rats. Delayed C1-inhibitor treatment still improved clinical outcome. Blood-brain barrier damage, edema formation, and inflammation were significantly lower compared with controls. Moreover, C1-inhibitor showed strong antithrombotic effects. CONCLUSIONS: C1-inhibitor is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Subject(s)
Anti-Inflammatory Agents , Brain Ischemia/prevention & control , Complement C1 Inhibitor Protein/therapeutic use , Fibrinolytic Agents , Reperfusion Injury/prevention & control , Animals , Blood-Brain Barrier/drug effects , Blotting, Western , Brain Edema/drug therapy , Brain Edema/pathology , Brain Ischemia/pathology , Female , Immunohistochemistry , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neurologic Examination , Rats , Real-Time Polymerase Chain Reaction , Reperfusion Injury/pathology , Sex Characteristics , Treatment Outcome
13.
Clin Transl Sci ; 15(3): 709-720, 2022 03.
Article in English | MEDLINE | ID: mdl-34811931

ABSTRACT

Factor XII (FXII) is a serine protease involved in multiple cascades, including the kallikrein-kinin system. It may play a role in diseases in which the downstream cascades are dysregulated, such as hereditary angioedema. Garadacimab (CSL312) is a first-in-class, fully human, monoclonal antibody targeting activated FXII (FXIIa). We describe how translational pharmacokinetic (PK) and pharmacodynamic (PD) modeling enabled dose selection for the phase I, first-in-human trial of garadacimab. The PK/PD data used for modeling were derived from preclinical PK/PD and safety studies. Garadacimab plasma concentrations rose with increasing dose, and clear dose-related PD effects were observed (e.g., a mechanism-based prolongation of activated partial thromboplastin time). The PK/PD profile from cynomolgus monkeys was used to generate minimal physiologically-based pharmacokinetic (mPBPK) models with target-mediated drug disposition (TMDD) for data prediction in cynomolgus monkeys. These models were later adapted for prediction of human data to establish dose selection. Based on the final mPBPK model with TMDD and assuming a weight of 70 kg for an adult human, a minimal inhibition (<10%) of FXIIa with a starting dose of 0.1 mg/kg garadacimab and a near maximal inhibition (>95%) at 10 mg/kg garadacimab were predicted. The phase I study is complete, and data on exposure profiles and inhibition of FXIIa-mediated kallikrein activity observed in the trial support and validate these simulations. This emphasizes the utility and relevance of translational modeling and simulation in drug development.


Subject(s)
Angioedemas, Hereditary , Factor XIIa , Animals , Antibodies, Monoclonal/pharmacokinetics , Computer Simulation , Humans , Macaca fascicularis
14.
Thromb Haemost ; 122(2): 196-207, 2022 02.
Article in English | MEDLINE | ID: mdl-34619795

ABSTRACT

BACKGROUND: 3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases. METHODS: The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease-angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE-/- model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation. RESULTS: Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers. CONCLUSION: Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Aortic Aneurysm, Abdominal/prevention & control , Atherosclerosis/prevention & control , Factor XIIa/antagonists & inhibitors , Plaque, Atherosclerotic/metabolism , Animals , Aortic Aneurysm, Abdominal/epidemiology , Apolipoproteins E , Disease Models, Animal , Inflammation , Male , Mice
15.
J Neurosci ; 30(31): 10369-79, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20685980

ABSTRACT

Oligomers of the beta-amyloid (Abeta) peptide have been indicated in early neuropathologic changes in Alzheimer's disease. Here, we present a synthetic Abeta(20-42) oligomer (named globulomer) with a different conformation to monomeric and fibrillar Abeta peptide, enabling the generation of highly Abeta oligomer-specific monoclonal antibodies. The globulomer-derived antibodies specifically detect oligomeric but not monomeric or fibrillar Abeta in various Abeta preparations. The globulomer-specific antibody A-887755 was able to prevent Abeta oligomer binding and dynamin cleavage in primary hippocampal neurons and to reverse globulomer-induced reduced synaptic transmission. In amyloid precursor protein (APP) transgenic mice, vaccination with Abeta globulomer and treatment with A-887755 improved novel object recognition. The cognitive improvement is likely attributable to reversing a deficit in hippocampal synaptic spine density in APP transgenic mice as observed after treatment with A-887755. Our findings demonstrate that selective reduction of Abeta oligomers by immunotherapy is sufficient to normalize cognitive behavior and synaptic deficits in APP transgenic mice.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Antibodies, Monoclonal/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Analysis of Variance , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/cytology , Hippocampus/immunology , Immunoprecipitation , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/immunology , Rats , Rats, Wistar , Recognition, Psychology
16.
J Thromb Haemost ; 19(11): 2835-2840, 2021 11.
Article in English | MEDLINE | ID: mdl-34363738

ABSTRACT

BACKGROUND: Effective inhibition of thrombosis without generating bleeding risks is a major challenge in medicine. Accumulating evidence suggests that this can be achieved by inhibition of coagulation factor XII (FXII), as either its knock-out or inhibition in animal models efficiently reduced thrombosis without affecting normal hemostasis. Based on these findings, highly specific inhibitors for human FXII(a) are under development. However, currently, in vivo studies on their efficacy and safety are impeded by the lack of an optimized animal model expressing the specific target, that is, human FXII. OBJECTIVE: The primary objective of this study is to develop and functionally characterize a humanized FXII mouse model. METHODS: A humanized FXII mouse model was generated by replacing the murine with the human F12 gene (genetic knock-in) and tested it in in vitro coagulation assays and in in vivo thrombosis models. RESULTS: These hF12KI mice were indistinguishable from wild-type mice in all tested assays of coagulation and platelet function in vitro and in vivo, except for reduced expression levels of hFXII compared to human plasma. Targeting FXII by the anti-human FXIIa antibody 3F7 increased activated partial thromboplastin time dose-dependently and protected hF12KI mice in an arterial thrombosis model without affecting bleeding times. CONCLUSION: These data establish the newly generated hF12KI mouse as a powerful and unique model system for in vivo studies on anti-FXII(a) biologics, supporting the development of efficient and safe human FXII(a) inhibitors.


Subject(s)
Factor XII , Thrombosis , Animals , Blood Coagulation , Disease Models, Animal , Factor XII/genetics , Hemostasis , Mice , Thrombosis/drug therapy , Thrombosis/genetics
17.
J Thromb Haemost ; 18(12): 3194-3202, 2020 12.
Article in English | MEDLINE | ID: mdl-32810892

ABSTRACT

BACKGROUND: Hemophilia B is caused by coagulation factor IX (FIX) deficiency. Recombinant fusion protein linking coagulation FIX with recombinant albumin (rIX-FP; Idelvion® ) is used for replacement therapy with an extended half-life. A previous quantitative whole-body autoradiography (QWBA) study investigating the biodistribution of rIX-FP indicated equal biodistribution, but more prolonged tissue retention compared with a marketed recombinant FIX product. OBJECTIVES: To complete and confirm the QWBA study data by directly measuring rIX-FP protein and activity levels in tissues following intravenous (i.v.) administration to normal rats and FIX-deficient (hemophilia B) mice. METHODS: After i.v. administration of rIX-FP at a dose of 2000 IU/kg, animals were euthanized at specific time points up to 72 hours postdosing. Subsequently, plasma and various tissues, which were selected based on the previous QWBA results, were harvested and analyzed for FIX antigen levels using an ELISA (both species) or an immunohistochemistry method (mice only), as well as for FIX activity levels (mice only) using a chromogenic assay. RESULTS: In rats, rIX-FP distributed extravascularly into all tissues analyzed (ie, liver, kidney, skin and knee) with peak antigen levels reached between 1 and 7 hours postdosing. In hemophilia B mice, rIX-FP tissue distribution was comparable to rats. FIX antigen levels correlated well with FIX activity readouts. CONCLUSIONS: Our results confirm QWBA data showing that rIX-FP distributes into relevant target tissues. Importantly, it was demonstrated that rIX-FP available in tissues retains its functional activity and can thus facilitate its therapeutic activity at sites of potential injury.


Subject(s)
Hemophilia B , Rodentia , Administration, Intravenous , Animals , Factor IX/metabolism , Half-Life , Hemophilia B/drug therapy , Mice , Rats , Recombinant Fusion Proteins/therapeutic use , Rodentia/metabolism , Tissue Distribution
18.
Neurobiol Dis ; 31(3): 342-54, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632280

ABSTRACT

Neural transplantation has been investigated experimentally and clinically for the purpose of developing new treatment options for intractable epilepsy. In the present study we assessed the anticonvulsant efficacy and safety of bilateral allotransplantation of genetically engineered striatal GABAergic rat cell lines into the substantia nigra pars reticulata (SNr). Rats with previously-established seizures, induced by amygdala kindling, were used as a model of temporal lobe epilepsy. Three cell lines were transplanted: (1) immortalized GABAergic cells (M213-2O) derived from embryonic rat striatum; (2) M213-2O cells (CL4) transfected with human GAD67 cDNA to obtain higher GABA synthesis than the parent cell line; and (3) control cells (121-1I), also derived from embryonic rat striatum, but which did not show GAD expression. A second control group received injections of medium alone. Transplantation of M213-2O cells into the SNr of kindled rats resulted in significant but transient anticonvulsant effects. Neither control cells nor medium induced anticonvulsant effects. Strong tissue reactions were, however, induced in the host brain of kindled but not of non-kindled rats, and only in animals that received grafts of genetically modified CL4 cells. These tissue reactions included graft rejection, massive infiltration of inflammatory immune cells, and gliosis. The anticonvulsant effect of M213-2O cells emphasizes the feasibility of local manipulations of seizures by intranigral transplantation of GABA-producing cells. On the other hand, the present data suggest that kindling-induced activation of microglia in the SNr can enhance immune reactions to transplanted cells. In this case, under conditions of further immunological stimulation by CL4 cells, transfected with a human cDNA, substantial immune reactions occurred. Thus, it appears that the condition of the host brain and the production of foreign proteins by transplanted cells have to be considered in estimating the risks of rejection of transplants into the brain.


Subject(s)
Brain Tissue Transplantation/methods , Epilepsy/metabolism , Epilepsy/surgery , Substantia Nigra/metabolism , Substantia Nigra/surgery , gamma-Aminobutyric Acid/biosynthesis , Animals , Brain Tissue Transplantation/adverse effects , Cell Line, Transformed , Disease Models, Animal , Epilepsy/physiopathology , Female , Genetic Therapy/methods , Glutamate Decarboxylase/genetics , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/prevention & control , Humans , Kindling, Neurologic/metabolism , Microglia/immunology , Neural Inhibition/physiology , Neurons/cytology , Neurons/metabolism , Neurons/transplantation , Rats , Rats, Wistar , Risk Assessment , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Substantia Nigra/physiopathology , Transfection/methods , Treatment Outcome , Up-Regulation/genetics
19.
Thromb Haemost ; 117(1): 176-187, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27786338

ABSTRACT

Atherosclerosis is considered a chronic inflammatory disease of the vessel wall. Coagulation pathways and immune responses contribute to disease development. The role of coagulation factor XII (FXII) in vascular inflammation, however, remains controversial. We here investigated the function of FXII in atherosclerosis using apolipoprotein E and FXII-deficient (F12-/-Apoe-/-) mice. Compared to F12+/+Apoe-/- controls, atherosclerotic lesion formation was reduced in F12-/-Apoe-/- mice. This was associated with a decrease in serum interleukin (IL)-1ß and IL-12 levels and reduced expression of pro-inflammatory cytokines in the aorta in atherosclerotic F12-/-Apoe-/- mice, as well as diminished Th1-cell differentiation in the aorta, blood, and lymphoid organs. No changes in circulating bradykinin, thrombin-antithrombin-complexes or plasminogen were observed. Mechanistically, activated FXII (FXIIa) was revealed to directly induce bone marrow-derived macrophages to secrete pro-inflammatory cytokines, including tumour necrosis factor-α, IL-1ß, IL-12, and IL-6. Exposure of bone marrow-derived antigen presenting cells to FXIIa similarly induced pro-inflammatory cytokines, and an enhanced capacity to trigger antigen-specific interferon γ-production in CD4+ T cells. Notably, bone-marrow derived macrophages were capable of directly activating FXII. Moreover, the induction of cytokine expression by FXIIa in macrophages occurred independently of FXII protease enzymatic activity and was decreased upon phospholipase C treatment, suggesting urokinase-type plasminogen activator receptor (uPAR) to confer FXIIa-induced cell signalling. These data reveal FXII to play an important role in atherosclerotic lesion formation by functioning as a strong inducer of pro-inflammatory cytokines in antigen-presenting cells. Targeting of FXII may thus be a promising approach for treating cardiovascular disease.


Subject(s)
Antigen-Presenting Cells/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cytokines/metabolism , Factor XII Deficiency/metabolism , Factor XII/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Animals , Antigen-Presenting Cells/immunology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Cell Proliferation , Cytokines/immunology , Disease Models, Animal , Factor XII/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/genetics , Factor XII Deficiency/immunology , Factor XIIa/genetics , Factor XIIa/metabolism , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Lymphocyte Activation , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , Receptors, Urokinase Plasminogen Activator/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors
20.
PLoS One ; 11(8): e0161826, 2016.
Article in English | MEDLINE | ID: mdl-27561008

ABSTRACT

In relation to the recent trials of Intravenous Immunoglobulin (IVIG) in Alzheimer's Disease (AD) it was demonstrated that different IgG preparations contain varying amounts of natural anti-amyloid ß (Aß) antibodies as measured by ELISA. We therefore investigated the relevance of ELISA data for measuring low-affinity antibodies, such as anti-Aß. We analysed the binding of different commercial Immunoglobulin G (IgG) preparations to Aß, actin and tetanus toxoid in different binding assays to further investigate the possible cause for observed differences in binding to Aß and actin between different IgG preparations. We show that the differences of commercial IgG preparations in binding to Aß and actin in ELISA assays are artefactual and only evident in in vitro binding assays. In functional assays and in vivo animal studies the different IVIG preparations exhibited very similar potency. ELISA data alone are not appropriate to analyse and rank the binding capacity of low-affinity antibodies to Aß or other endogenous self-antigens contained in IgG preparations. Additional analytical methods should be adopted to complement ELISA data.


Subject(s)
Antigens/immunology , Autoantigens/immunology , Immunoglobulin G/immunology , Immunoglobulins, Intravenous/immunology , Actins/immunology , Actins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibody Affinity/drug effects , Antibody Affinity/immunology , Antigens/metabolism , Autoantigens/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Herpesvirus 3, Human/immunology , Herpesvirus 3, Human/metabolism , Humans , Immunoglobulin G/metabolism , Immunoglobulins, Intravenous/pharmacology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/drug effects , Protein Binding/immunology , Rats , Tetanus Toxoid/immunology , Tetanus Toxoid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL