Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.096
Filter
1.
Blood ; 142(12): 1071-1081, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37294924

ABSTRACT

Rebalance of coagulation and anticoagulation to achieve a hemostatic effect has recently gained attention as an alternative therapeutic strategy for hemophilia. We engineered a humanized chimeric antibody, SR604, based on a previously published murine antibody, HAPC1573, which selectively blocks the anticoagulant activity of human activated protein C (APC). SR604 effectively blocked the anticoagulation activities of APC in human plasma deficient in various coagulation factors in vitro with affinities ∼60 times greater than that of HAPC1573. SR604 exhibited prophylactic and therapeutic efficacy in the tail-bleeding and knee-injury models of hemophilia A and B mice expressing human APC (humanized hemophilic mice). SR604 did not interfere with the cytoprotection and endothelial barrier function of APC, nor were there obvious toxicity effects in humanized hemophilic mice. Pharmacokinetic study showed a high bioavailability (106%) of subcutaneously injected SR604 in cynomolgus monkeys. These results demonstrate that SR604 is expected to be a safe and effective therapeutic and/or prophylactic agent with a prolonged half-life for patients with congenital factor deficiencies including hemophilia A and B.


Subject(s)
Hemophilia A , Protein C , Humans , Mice , Animals , Protein C/therapeutic use , Hemophilia A/drug therapy , Disease Models, Animal , Blood Coagulation , Anticoagulants/therapeutic use
2.
Circ Res ; 130(2): 252-272, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34930019

ABSTRACT

BACKGROUND: APC (activated protein C) is a plasma serine protease with anticoagulant and anti-inflammatory activities. EPCR (Endothelial protein C receptor) is associated with APC's activity and mediates its downstream signaling events. APC exerts cardioprotective effects during ischemia and reperfusion (I/R). This study aims to characterize the role of the APC-EPCR axis in ischemic insults in aging. METHODS: Young (3-4 months) and aged (24-26 months) wild-type C57BL/6J mice, as well as EPCR point mutation (EPCRR84A/R84A) knockin C57BL/6J mice incapable of interaction with APC and its wild type of littermate C57BL/6J mice, were subjected to I/R. Wild-type APC, signaling-selective APC-2Cys, or anticoagulant-selective APC-E170A were administrated before reperfusion. RESULTS: The results demonstrated that cardiac I/R reduces APC activity, and the APC activity was impaired in the aged versus young hearts possibly attributable to the declined EPCR level with aging. Serum EPCR measurement showed that I/R triggered the shedding of membrane EPCR into circulation, while administration of APC attenuated the I/R-induced EPCR shedding in both young and aged hearts. Subsequent echocardiography showed that APC and APC-2Cys but not APC-E170A ameliorated cardiac dysfunction during I/R in both young and aged mice. Importantly, APC elevated the resistance of the aged heart to ischemic insults through stabilizing EPCR. However, all these cardioprotective effects of APC were blunted in the EPCRR84A/R84A mice versus its wild-type littermates. The ex vivo working heart and metabolomics results demonstrated that AMPK (AMP-activated protein kinase) mediates acute adaptive response while AKT (protein kinase B) is involved in chronic metabolic programming in the hearts with APC treatment. CONCLUSIONS: I/R stress causes shedding of the membrane EPCR in the heart, and administration of APC prevents I/R-induced cardiac EPCR shedding that is critical for limiting cardiac damage in aging.


Subject(s)
Aging/metabolism , Endothelial Protein C Receptor/metabolism , Myocardial Reperfusion Injury/metabolism , Protein C/metabolism , Animals , Cardiotonic Agents/therapeutic use , Endothelial Protein C Receptor/blood , Female , Heart/growth & development , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/drug therapy , Myocardium/metabolism , Protein C/therapeutic use
3.
Vox Sang ; 119(3): 193-202, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38018260

ABSTRACT

BACKGROUND AND OBJECTIVES: Deficiencies of protein C (PC) or protein S (PS) are rare diseases, characterized by mutations in the PC or PS genes, which encode plasma serine proteases with anti-coagulant activity. Severe PC or PS deficiencies manifest in early life as neonatal purpura fulminans, a life-threatening heamorrhagic condition requiring immediate treatment. First-line treatment involves replacement therapy, followed by maintenance with anti-coagulants. Replacement therapy with specific protein concentrates is currently only limited to PC, and therefore, a PC + PS concentrate represents a useful addition to therapeutic options, particularly for severe PS deficiency. Further, the production of a PC + PS concentrate from unused plasma fractionation intermediates would impact favourably on manufacturing costs, and consequently therapy prices for patients and health systems. MATERIALS AND METHODS: Several chromatographic runs were performed on the same unused plasma fractionation intermediates using different supports to obtain a PC/PS concentrate. The best chromatographic mediums were chosen, in terms of specific activity and recovery. A full process of purification including virus inactivation/removal and lyophilization steps was set up. RESULTS: The final freeze-dried product had a mean PC concentration of 47.75 IU/mL with 11% of PS, and a mean specific activity of 202.5 IU/mg protein, corresponding to over 12,000-fold purification from plasma. CONCLUSION: The development of a novel concentrated PC/PS mixture obtained from a waste fraction of other commercial products could be used for its potential therapeutic role in the management of neonatal purpura fulminans pathology.


Subject(s)
Protein C Deficiency , Purpura Fulminans , Infant, Newborn , Humans , Purpura Fulminans/drug therapy , Purpura Fulminans/genetics , Protein C Deficiency/drug therapy , Protein C/analysis , Protein C/therapeutic use , Protein S , Plasma/chemistry
4.
Wound Repair Regen ; 32(1): 90-103, 2024.
Article in English | MEDLINE | ID: mdl-38155595

ABSTRACT

Various preclinical and clinical studies have demonstrated the robust wound healing capacity of the natural anticoagulant activated protein C (APC). A bioengineered APC variant designated 3K3A-APC retains APC's cytoprotective cell signalling actions with <10% anticoagulant activity. This study was aimed to provide preclinical evidence that 3K3A-APC is efficacious and safe as a wound healing agent. 3K3A-APC, like wild-type APC, demonstrated positive effects on proliferation of human skin cells (keratinocytes, endothelial cells and fibroblasts). Similarly it also increased matrix metollaproteinase-2 activation in keratinocytes and fibroblasts. Topical 3K3A-APC treatment at 10 or 30 µg both accelerated mouse wound healing when culled on Day 11. And at 10 µg, it was superior to APC and had half the dermal wound gape compared to control. Further testing was conducted in excisional porcine wounds due to their congruence to human skin. Here, 3K3A-APC advanced macroscopic healing in a dose-dependent manner (100, 250 and 500 µg) when culled on Day 21. This was histologically corroborated by greater collagen maturity, suggesting more advanced remodelling. A non-interference arm of this study found no evidence that topical 3K3A-APC caused either any significant systemic side-effects or any significant leakage into the circulation. However the female pigs exhibited transient and mild local reactions after treatments in week three, which did not impact healing. Overall these preclinical studies support the hypothesis that 3K3A-APC merits future human wound studies.


Subject(s)
Endothelial Cells , Protein C , Female , Humans , Animals , Mice , Swine , Protein C/pharmacology , Protein C/metabolism , Protein C/therapeutic use , Endothelial Cells/metabolism , Wound Healing , Fibrinolytic Agents/therapeutic use , Anticoagulants/pharmacology , Anticoagulants/therapeutic use
5.
Circ Res ; 128(4): 513-529, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33353373

ABSTRACT

RATIONALE: While thrombin is the key protease in thrombus formation, other coagulation proteases, such as fXa (factor Xa) or aPC (activated protein C), independently modulate intracellular signaling via partially distinct receptors. OBJECTIVES: To study the differential effects of fXa or fIIa (factor IIa) inhibition on gene expression and inflammation in myocardial ischemia-reperfusion injury. METHODS AND RESULTS: Mice were treated with a direct fIIa inhibitor (fIIai) or direct fXa inhibitor (fXai) at doses that induced comparable anticoagulant effects ex vivo and in vivo (tail-bleeding assay and FeCl3-induced thrombosis). Myocardial ischemia-reperfusion injury was induced via left anterior descending ligation. We determined infarct size and in vivo aPC generation, analyzed gene expression by RNA sequencing, and performed immunoblotting and ELISA. The signaling-only 3K3A-aPC variant and inhibitory antibodies that blocked all or only the anticoagulant function of aPC were used to determine the role of aPC. Doses of fIIai and fXai that induced comparable anticoagulant effects resulted in a comparable reduction in infarct size. However, unbiased gene expression analyses revealed marked differences, including pathways related to sterile inflammation and inflammasome regulation. fXai but not fIIai inhibited sterile inflammation by reducing the expression of proinflammatory cytokines (IL [interleukin]-1ß, IL-6, and TNFα [tumor necrosis factor alpha]), as well as NF-κB (nuclear factor kappa B) and inflammasome activation. This anti-inflammatory effect was associated with reduced myocardial fibrosis 28 days post-myocardial ischemia-reperfusion injury. Mechanistically, in vivo aPC generation was higher with fXai than with fIIai. Inhibition of the anticoagulant and signaling properties of aPC abolished the anti-inflammatory effect associated with fXai, while inhibiting only the anticoagulant function of aPC had no effect. Combining 3K3A-aPC with fIIai reduced the inflammatory response, mimicking the fXai-associated effect. CONCLUSIONS: We showed that specific inhibition of coagulation via direct oral anticoagulants had differential effects on gene expression and inflammation, despite comparable anticoagulant effects and infarct sizes. Targeting individual coagulation proteases induces specific cellular responses unrelated to their anticoagulant effect.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Factor Xa Inhibitors/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Protein C/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Factor Xa Inhibitors/pharmacology , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Protein C/pharmacology
6.
Int J Mol Sci ; 24(13)2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37445820

ABSTRACT

3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with cytoprotective properties and reduced bleeding risks. We studied the potential use of 3K3A-APC as a multi-target therapeutic option for choroidal neovascularization (CNV), a common cause of vision loss in age-related macular degeneration. CNV was induced by laser photocoagulation in a murine model, and 3K3A-APC was intravitreally injected. The impact of 3K3A-APC treatment on myeloid and microglia cell activation and recruitment and on NLRP3 inflammasome, IL-1ß, and VEGF levels was assessed using cryosection, retinal flat-mount immunohistochemistry and vascular imaging. Additionally, we evaluated the use of fluorescein angiography as a surrogate marker for in vivo evaluation of the efficacy of 3K3A-APC treatment against leaking CNV lesions. Our results demonstrated that 3K3A-APC treatment significantly reduced the accumulation and activation of myeloid cells and microglia in the CNV area and decreased the NLRP3 and IL-1ß levels at the CNV site and the surrounding retina. Furthermore, 3K3A-APC treatment resulted in leakage regression and CNV growth suppression. These findings indicate that the anti-inflammatory activities of 3K3A-APC contribute to CNV inhibition. Our study suggests the potential use of 3K3A-APC as a novel multi-target treatment for CNV.


Subject(s)
Choroidal Neovascularization , Protein C , Mice , Animals , Protein C/pharmacology , Protein C/therapeutic use , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Vascular Endothelial Growth Factor A , Retina/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Mice, Inbred C57BL
7.
J Neuroinflammation ; 19(1): 138, 2022 Jun 11.
Article in English | MEDLINE | ID: mdl-35690769

ABSTRACT

BACKGROUND: Inflammation and coagulation are linked and pathogenic in neuroinflammatory diseases. Protease-activated receptor 1 (PAR1) can be activated both by thrombin, inducing increased inflammation, and activated protein C (aPC), inducing decreased inflammation. Modulation of the aPC-PAR1 pathway may prevent the neuroinflammation associated with PAR1 over-activation. METHODS: We synthesized a group of novel molecules based on the binding site of FVII/aPC to the endothelial protein C receptor (EPCR). These molecules modulate the FVII/aPC-EPCR pathway and are therefore named FEAMs-Factor VII, EPCR, aPC Modulators. We studied the molecular and behavioral effects of a selected FEAM in neuroinflammation models in-vitro and in-vivo. RESULTS: In a lipopolysaccharide (LPS) induced in-vitro model, neuroinflammation leads to increased thrombin activity compared to control (2.7 ± 0.11 and 2.23 ± 0.13 mU/ml, respectively, p = 0.01) and decreased aPC activity (0.57 ± 0.01 and 1.00 ± 0.02, respectively, p < 0.0001). In addition, increased phosphorylated extracellular regulated kinase (pERK) (0.99 ± 0.13, 1.39 ± 0.14, control and LPS, p < 0.04) and protein kinase B (pAKT) (1.00 ± 0.09, 2.83 ± 0.81, control and LPS, p < 0.0002) levels indicate PAR1 overactivation, which leads to increased tumor necrosis factor-alpha (TNF-α) level (1.00 ± 0.04, 1.35 ± 0.12, control and LPS, p = 0.02). In a minimal traumatic brain injury (mTBI) induced neuroinflammation in-vivo model in mice, increased thrombin activity, PAR1 activation, and TNF-α levels were measured. Additionally, significant memory impairment, as indicated by a lower recognition index in the Novel Object Recognition (NOR) test and Y-maze test (NOR: 0.19 ± 0.06, -0.07 ± 0.09, p = 0.03. Y-Maze: 0.50 ± 0.03, 0.23 ± 0.09, p = 0.02 control and mTBI, respectively), as well as hypersensitivity by hot-plate latency (16.6 ± 0.89, 12.8 ± 0.56 s, control and mTBI, p = 0.01), were seen. FEAM prevented most of the molecular and behavioral negative effects of neuroinflammation in-vitro and in-vivo, most likely through EPCR-PAR1 interactions. CONCLUSION: FEAM is a promising tool to study neuroinflammation and a potential treatment for a variety of neuroinflammatory diseases.


Subject(s)
Protein C , Receptor, PAR-1 , Animals , Endothelial Protein C Receptor/metabolism , Factor VII/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , Neuroinflammatory Diseases , Protein C/metabolism , Protein C/therapeutic use , Receptor, PAR-1/metabolism , Signal Transduction , Thrombin/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Chem Biodivers ; 19(10): e202200494, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36198620

ABSTRACT

Biofilm-producing Staphylococcus aureus (SA) strains are frequently found in medical environments, from surgical/ wound sites, medical devices. These biofilms reduce the efficacy of applied antibiotics during the treatment of several infections, such as cystic fibrosis, endocarditis, or urinary tract infections. Thus, the development of potential therapeutic agents to destroy the extra protective biofilm layers or to inhibit the biofilm-producing enzymes is urgently needed. Advanced and cost-effective bioinformatics tools are advantageous in locating and speeding up the selection of antibiofilm candidates. Based on the potential drug characteristics, we have selected one-hundred thirty-three antibacterial peptides derived from insects to assess for their antibiofilm potency via molecular docking against five putative biofilm formation and regulated target enzymes: the staphylococcal accessory regulator A or SarA (PDB ID: 2FRH), 4,4'-diapophytoene synthase or CrtM (PDB ID: 2ZCQ), clumping factor A or ClfA (PDB ID: 1N67) and serine-aspartate repeat protein C or SdrC (PDB ID: 6LXH) and sortase A or SrtA (PDB ID: 1T2W) of SA bacterium. In this study, molecular docking was performed using HPEPDOCK and HDOCK servers, and molecular interactions were examined using BIOVIA Discovery Studio Visualizer-2019. The docking score (kcal/mol) range of five promising antibiofilm peptides against five targets was recorded as follows: diptericin A (-215.52 to -303.31), defensin (-201.11 to -301.92), imcroporin (-212.08 to -287.64), mucroporin (-228.72 to -286.76), apidaecin II (-203.90 to -280.20). Among these five, imcroporin and mucroporin were 13 % each, while defensin contained only 1 % of positive net charged residues (Arg+Lys) projected through ProtParam and NetWheels tools. Similarly, imcroporin, mucroporin and apidaecin II were 50 %, while defensin carried 21.05 % of hydrophobic residues predicted by the tool PEPTIDE. 2.0. Most of the peptides exhibited potential characteristics to inhibit S. aureus-biofilm formation via disrupting the cell membrane and cytoplasmic integrity. In summary, the proposed hypothesis can be considered a cost-effective platform for selecting the most promising bioactive drug candidates within a limited timeframe with a greater chance of success in experimental and clinical studies.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Molecular Docking Simulation , Protein C/pharmacology , Protein C/therapeutic use , Aspartic Acid/pharmacology , Aspartic Acid/therapeutic use , Staphylococcal Infections/drug therapy , Biofilms , Anti-Bacterial Agents/pharmacology , Defensins/pharmacology , Defensins/therapeutic use , Insecta , Serine/pharmacology , Serine/therapeutic use , Microbial Sensitivity Tests
9.
Int J Mol Sci ; 23(15)2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35955419

ABSTRACT

The vascular endothelium has several important functions, including hemostasis. The homeostasis of hemostasis is based on a fine balance between procoagulant and anticoagulant proteins and between fibrinolytic and antifibrinolytic ones. Coagulopathies are characterized by a mutation-induced alteration of the function of certain coagulation factors or by a disturbed balance between the mechanisms responsible for regulating coagulation. Homeostatic therapies consist in replacement and nonreplacement treatments or in the administration of antifibrinolytic agents. Rebalancing products reestablish hemostasis by inhibiting natural anticoagulant pathways. These agents include monoclonal antibodies, such as concizumab and marstacimab, which target the tissue factor pathway inhibitor; interfering RNA therapies, such as fitusiran, which targets antithrombin III; and protease inhibitors, such as serpinPC, which targets active protein C. In cases of thrombophilia (deficiency of protein C, protein S, or factor V Leiden), treatment may consist in direct oral anticoagulants, replacement therapy (plasma or recombinant ADAMTS13) in cases of a congenital deficiency of ADAMTS13, or immunomodulators (prednisone) if the thrombophilia is autoimmune. Monoclonal-antibody-based anti-vWF immunotherapy (caplacizumab) is used in the context of severe thrombophilia, regardless of the cause of the disorder. In cases of disseminated intravascular coagulation, the treatment of choice consists in administration of antifibrinolytics, all-trans-retinoic acid, and recombinant soluble human thrombomodulin.


Subject(s)
Factor V/metabolism , Thrombophilia , von Willebrand Factor , Anticoagulants , Endothelium, Vascular/metabolism , Factor VIII/genetics , Factor VIII/therapeutic use , Homeostasis , Humans , Protein C/therapeutic use , Thrombophilia/genetics , von Willebrand Factor/metabolism
10.
Int J Mol Sci ; 23(22)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36430674

ABSTRACT

3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with pleiotropic cytoprotective properties albeit without the bleeding risks. The anti-inflammatory activities of 3K3A-APC were demonstrated in multiple preclinical injury models, including various neurological disorders. We determined the ability of 3K3A-APC to inhibit ocular inflammation in a murine model of lipopolysaccharide (LPS)-induced uveitis. Leukocyte recruitment, microglia activation, NLRP3 inflammasome and IL-1ß levels were assessed using flow cytometry, retinal cryosection histology, retinal flatmount immunohistochemistry and vascular imaging, with and without 3K3A-APC treatment. LPS triggered robust inflammatory cell recruitment in the posterior chamber. The 3K3A-APC treatment significantly decreased leukocyte numbers and inhibited leukocyte extravasation from blood vessels into the retinal parenchyma to a level similar to controls. Resident microglia, which underwent an inflammatory transition following LPS injection, remained quiescent in eyes treated with 3K3A-APC. An inflammation-associated increase in retinal thickness, observed in LPS-injected eyes, was diminished by 3K3A-APC treatment, suggesting its clinical relevancy. Finally, 3K3A-APC treatment inhibited inflammasome activation, determined by lower levels of NLRP3 and its downstream effector IL-1ß. Our results highlight the anti-inflammatory properties of 3K3A-APC in ocular inflammation and suggest its potential use as a novel treatment for retinal diseases associated with inflammation.


Subject(s)
Eye Diseases , Inflammasomes , Protein C , Animals , Mice , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein C/pharmacology , Protein C/therapeutic use , Eye Diseases/drug therapy , Eye Diseases/pathology
11.
Blood ; 134(9): 776-781, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31243040

ABSTRACT

Soluble thrombomodulin plasma concentrations are elevated in steroid-resistant graft-versus-host disease (GVHD), implying endothelial hypofunctioning for thrombomodulin-dependent generation of activated protein C's (APC) anticoagulant, anti-inflammatory, and antiapoptotic functions. Recombinant thrombomodulin or APC administration decreases acute GVHD, manifested by intense inflammation and tissue destruction. Here, we administered recombinant murine wild-type (WT) APC to mice with established chronic GVHD (cGVHD), a less-inflammatory autoimmune-like disease. WT APC normalized bronchiolitis obliterans-induced pulmonary dysfunction. Signaling-selective APC variants (3A-APC [APC with lysine 191-193 replaced with 3 alanines] or 5A-APC [APC with lysine 191-193 replaced with 3 alanines and arginine 229/230 replaced with 2 alanines]) with normal cytoprotective properties, but greatly reduced anticoagulant activity, provided similar results. Mechanistically, WT APC and signaling-selective variants reduced T follicular helper cells, germinal center formation, immunoglobulin, and collagen deposition. WT APC can potentially cleave protease-activated receptor 1 (PAR1) at Arg41 or Arg46, the latter causing anti-inflammatory signaling. cGVHD was reduced in recipients of T cells from WT PAR1 or mutated Gln41-PAR1 donors but not from mutated Gln46-PAR1 donors. These data implicate donor T-cell APC-induced noncanonical cleavage at Arg46-PAR1, which is known to confer cytoprotective and anti-inflammatory activities. Together, these data indicate that APC anticoagulant activity is dispensable, whereas anti-inflammatory signaling and cytoprotective cell signaling by APC are essential. Because a phase 2 ischemic stroke clinical trial did not raise any safety issues for 3A-APC treatment, our studies provide a foundational platform for testing in clinical cGVHD therapy.


Subject(s)
Graft vs Host Disease/drug therapy , Protein C/therapeutic use , Receptor, PAR-1/metabolism , T-Lymphocytes/drug effects , Animals , Chronic Disease , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Recombinant Proteins/therapeutic use , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
12.
Graefes Arch Clin Exp Ophthalmol ; 259(10): 2919-2927, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33893866

ABSTRACT

PURPOSE: Our previous 1-year pilot study evaluated the efficacy of intravitreally injected activated protein C (APC) in 10 eyes with ischemic central retinal vein occlusion (CRVO). The reperfusion of the areas of retinal nonperfusion (RNP) exceeded 50% of the baseline in five (50%) eyes 1 year after the APC injection. The current study evaluated the long-term efficacy and safety of intravitreal APC. METHODS: The 10 eyes in the pilot study were included in this study. Other treatments were administered at the physicians' discretion after the pilot study. We evaluated visual acuity (VA), central retinal thickness (CRT) and perfusion status, and adverse events and severity over the long term. RESULTS: The median follow-up was 60 months (range, 48-68 months). Compared with baseline, the post-treatment VA improved significantly (P < 0.001) from 1.39 to 1.06 logarithm of the minimum angle of resolution. The CRT improved significantly (P < 0.001) from 1090 to 195 µm at the last visit. The RNP areas decreased from an average 29.7 disc areas (DAs) at baseline to an average 16.5 DAs at the last examination (mean, 40 ± 6.5 months after the first APC treatment). No adverse events were related to intravitreal APC. CONCLUSION: No complications were associated with intravitreal APC, the clinical course improved, and improved RNP was maintained for the long term, suggesting that intravitreal APC may be an alternative treatment for CRVO.


Subject(s)
Macular Edema , Retinal Vein Occlusion , Angiogenesis Inhibitors/therapeutic use , Follow-Up Studies , Humans , Intravitreal Injections , Macular Edema/drug therapy , Pilot Projects , Protein C/therapeutic use , Retinal Vein Occlusion/diagnosis , Retinal Vein Occlusion/drug therapy , Tomography, Optical Coherence , Treatment Outcome
13.
Curr Opin Hematol ; 26(1): 41-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30451721

ABSTRACT

PURPOSE OF REVIEW: The serine protease activated protein C (aPC) was initially characterized as an endogenous anticoagulant, but in addition conveys anti-inflammatory, barrier-protective, and pro cell-survival functions. Its endogenous anticoagulant function hampered the successful and continuous implantation of aPC as a therapeutic agent in septic patients. However, it became increasingly apparent that aPC controls cellular function largely independent of its anticoagulant effects through cell-specific and context-specific receptor complexes and intracellular signaling pathways. The purpose of this review is to outline the mechanisms of aPC-dependent cell signaling and its intracellular molecular targets. RECENT FINDINGS: With the advent of new therapeutic agents either modulating directly and specifically the activity of coagulation proteases or interfering with protease-activated receptor signaling a better understanding not only of the receptor mechanisms but also of the intracellular signaling mechanisms controlled by aPC in a disease-specific and context-specific fashion, is required to tailor new therapeutic approaches based on aPC's anti-inflammatory, barrier-protective, and pro cell-survival functions. SUMMARY: This review summarizes recent insights into the intracellular signaling pathways controlled by aPC in a cell-specific and context-specific fashion. We focus on aPC-mediated barrier protection, inhibition of inflammation, and cytoprotecting within this review.


Subject(s)
Anticoagulants , Protein C , Sepsis , Signal Transduction/drug effects , Animals , Anticoagulants/metabolism , Anticoagulants/therapeutic use , Humans , Protein C/metabolism , Protein C/therapeutic use , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/pathology
14.
Exp Eye Res ; 186: 107695, 2019 09.
Article in English | MEDLINE | ID: mdl-31201804

ABSTRACT

Activated protein C (APC) exerts diverse cell signaling pathways which results in multiple distinct cytoprotective actions. These include anti-apoptotic and anti-inflammatory activities and stabilization of endothelial and epithelial barriers. We studied the ability of APC to inhibit the leakage and the growth of newly formed as well as pre-existing choroidal neovascularization (CNV) and examined the ability of APC to stabilize the Retinal Pigmented Epithelium (RPE). We explored the contribution of Tie2 receptor to the protective effects of APC. CNV was induced by laser photocoagulation in C57BL/6J mice. APC was injected intravitreally immediately or 7 days after CNV induction. Neovascularization was evaluated on RPE-choroidal flatmounts using FITC-dextran perfusion and CD31 immunofluorescence. CNV leakage was measured by fluorescein angiography (FA). The ability of APC to stabilize the RPE barrier was evaluated in-vitro by dextran permeability and zonula occludens 1 (ZO1) immunostaining. Tie2 blocking was induced in-vivo by intraperitoneal injection of Tie2 kinase inhibitor and in-vitro by incubation with anti Tie2 antibodies. APC treatment dramatically inhibited the generation of newly formed CNV leakage sites and reversed leakage in 85% of the pre-existing CNV leaking sites. In RPE cell culture, APC induced translocation of ZO1 to the cell membrane, accompanied by reduction in permeability of the monolayer. Inhibition of Tie2 significantly decreased APC protective activities in both the mouse model and the RPE cell culture. Our results show that APC treatment significantly inhibits the leakage and growth of newly formed, as well as pre-existing CNV, and its protective activities are partially mediated via the Tie2 receptor. The data suggest that APC should be further investigated as a possible effective treatment for CNV.


Subject(s)
Anti-Infective Agents/therapeutic use , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Protein C/therapeutic use , Animals , Capillary Permeability/drug effects , Cell Membrane Permeability , Choroid/blood supply , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/physiopathology , Dose-Response Relationship, Drug , Fluorescein Angiography , Fluorescent Antibody Technique, Indirect , Humans , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/therapeutic use , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Zonula Occludens-1 Protein/metabolism
15.
J Thromb Thrombolysis ; 45(2): 315-318, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29270858

ABSTRACT

Purpura fulminans in neonates is a rapidly progressive thrombotic disorder manifesting as hemorrhagic skin infarction and disseminated intravascular coagulation. Being inherited in an autosomal dominant manner, it is a medical emergency. Clinical presentations of patients may vary depending on the genetic mutations. Retinal and intracranial hemorrhages are the worst clinical scenarios with persistent morbidity. During acute phase, fresh frozen plasma, protein C concentrates and anticoagulant therapy should be administered rapidly. Here we report a patient with homozygous protein C deficiency.


Subject(s)
Protein C Deficiency/therapy , Diagnosis, Differential , Heparin, Low-Molecular-Weight/therapeutic use , Homozygote , Humans , Infant, Newborn , Plasma , Protein C/administration & dosage , Protein C/therapeutic use , Purpura Fulminans
16.
Blood ; 126(7): 915-9, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26084674

ABSTRACT

Activated protein C (APC) is an anticoagulant protease that initiates cell signaling via protease-activated receptor 1 (PAR1) to regulate vascular integrity and inflammatory response. In this study, a recombinant APC variant (APC(N329Q)) mimicking the naturally occurring APC-ß plasma glycoform was found to exhibit superior PAR1 proteolysis at a cleavage site that selectively mediates cytoprotective signaling. APC(N329Q) also enhanced integrin αMß2-dependent PAR1 proteolysis to exert significantly improved antiinflammatory activity on macrophages compared with wild-type APC. Recent therapeutic applications of recombinant APC in ischemic stroke models have used APC variants with limited anticoagulant activity to negate potential bleeding side effects. Using a mouse model of ischemic stroke and late t-PA intervention, the neuroprotective activity of a murine APC variant with limited anticoagulant activity (mAPC(PS)) was compared with an identical APC variant except for the absence of glycosylation at the APC-ß sequon (mAPC(PS/N329Q)). Remarkably, mAPC(PS/N329Q) limited cerebral ischemic injury and reduced brain lesion volume significantly more effectively than mAPC(PS). Collectively, this study reveals the importance of APC glycosylation in controlling the efficacy of PAR1 proteolysis by APC and demonstrates the potential of novel APC variants with superior cytoprotective signaling function as enhanced therapeutic agents for the treatment of ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Protein C/metabolism , Receptor, PAR-1/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Brain Ischemia/therapy , Cathepsins/genetics , Cathepsins/metabolism , Disease Models, Animal , Endothelial Protein C Receptor , Genetic Variation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Oligosaccharides , Protein C/genetics , Protein C/therapeutic use , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/therapeutic use , Proteolysis , Receptor, PAR-1/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Signal Transduction
17.
Stat Med ; 36(8): 1334-1360, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27891631

ABSTRACT

Clinical trials target patients who are expected to benefit from a new treatment under investigation. However, the magnitude of the treatment benefit, if it exists, often depends on the patient baseline characteristics. It is therefore important to investigate the consistency of the treatment effect across subgroups to ensure a proper interpretation of positive study findings in the overall population. Such assessments can provide guidance on how the treatment should be used. However, great care has to be taken when interpreting consistency results. An observed heterogeneity in treatment effect across subgroups can arise because of chance alone, whereas true heterogeneity may be difficult to detect by standard statistical tests because of their low power. This tutorial considers issues related to subgroup analyses and their impact on the interpretation of findings of completed trials that met their main objectives. In addition, we provide guidance on the design and analysis of clinical trials that account for the expected heterogeneity of treatment effects across subgroups by establishing treatment benefit in a pre-defined targeted subgroup and/or the overall population. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Data Interpretation, Statistical , Randomized Controlled Trials as Topic , Aspirin/therapeutic use , Clopidogrel , Humans , Models, Statistical , Myocardial Infarction/prevention & control , Protein C/therapeutic use , Randomized Controlled Trials as Topic/methods , Recombinant Proteins/therapeutic use , Reproducibility of Results , Sepsis/drug therapy , Sepsis/mortality , Statistics as Topic , Stroke/prevention & control , Survival Analysis , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use , Treatment Outcome
18.
Arterioscler Thromb Vasc Biol ; 36(11): 2143-2151, 2016 11.
Article in English | MEDLINE | ID: mdl-27758767

ABSTRACT

APC (activated protein C), derived from the plasma protease zymogen, is antithrombotic and anti-inflammatory. In preclinical injury models, recombinant APC provides neuroprotection for multiple injuries, including ischemic stroke. APC acts directly on brain endothelial cells and neurons by initiating cell signaling that requires multiple receptors. Two or more major APC receptors mediate APC's neuroprotective cell signaling. When bound to endothelial cell protein C receptor, APC can cleave protease-activated receptor 1, causing biased cytoprotective signaling that reduces ischemia-induced injury. Pharmacological APC alleviates bleeding induced by tissue-type plasminogen activator in murine ischemic stroke studies. Remarkably, APC's signaling promotes neurogenesis. The signaling-selective recombinant variant of APC, 3K3A-APC, was engineered to lack most of the APC's anticoagulant activity but retain APC's cell signaling actions. Recombinant 3K3A-APC is in ongoing National Institutes of Health (NIH)-funded clinical trials for ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Intracranial Thrombosis/drug therapy , Neuroprotective Agents/therapeutic use , Protein C/therapeutic use , Recombinant Proteins/therapeutic use , Reperfusion Injury/prevention & control , Stroke/drug therapy , Thrombolytic Therapy/adverse effects , Animals , Antigens, CD/metabolism , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Protein C Receptor , Hemostasis/drug effects , Humans , Intracranial Thrombosis/metabolism , Intracranial Thrombosis/pathology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/adverse effects , Protein C/adverse effects , Receptor Cross-Talk/drug effects , Receptor, PAR-1/metabolism , Receptors, Cell Surface/agonists , Receptors, Cell Surface/metabolism , Recombinant Proteins/adverse effects , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Stroke/metabolism , Stroke/pathology
19.
Pediatr Blood Cancer ; 63(8): 1488-90, 2016 08.
Article in English | MEDLINE | ID: mdl-27138381

ABSTRACT

Subcutaneous (SC) protein C (PC) was used in a child with purpura fulminans secondary to severe congenital PC deficiency. For maintenance, PC 80-120 IU/kg, given over 60-90 min SC Q48hr, has been successful as a home therapy for more than 3 years. The treatment was monitored by measuring trough PC chromogenic activity (target ≥15%) and D-dimer levels. No change in clinical course was appreciated after discontinuing enoxaparin (and leaving the patient on prophylactic PC replacement alone). A significant discrepancy between clotting-based and chromogenic-based PC activity is shown.


Subject(s)
Protein C Deficiency/drug therapy , Protein C Deficiency/pathology , Protein C/genetics , Protein C/therapeutic use , Anticoagulants/therapeutic use , Enoxaparin/therapeutic use , Female , Humans , Infant, Newborn , Liver Transplantation , Protein C/administration & dosage
20.
Scand J Clin Lab Invest ; 76(1): 10-6, 2016.
Article in English | MEDLINE | ID: mdl-26403265

ABSTRACT

OBJECTIVES: Activated protein C (APC), an endogenous anticoagulant, has antithrombotic, fibrinolytic and anti-inflammatory properties. We recently conducted a controlled study (APCAP, activated protein C in severe acute pancreatitis) of APC treatment of patients with severe acute pancreatitis (SAP). Here we studied the effect of APC on the pivotal coagulation parameters of the surviving patients in the APCAP study. METHODS: The study consisted of 20 patients of whom 10 patients had received APC and 10 patients had received placebo. Coagulation parameters, physiological anticoagulants, thrombograms and circulating levels of IL-6 and CRP were determined on admission and at days 1, 3-4 and 6-7. RESULTS: During follow-up, the temporal levels of prothrombin time (PT) decreased and the temporal levels of thromboplastin time (TT) increased in placebo group (p< 0.001 for both), but not in APC group. The temporal levels of antithrombin (AT) increased less in APC group than in placebo group (p = 0.011). The shapes of the SAP patients' thrombograms were strongly deranged and were marginally affected by APC treatment. CONCLUSIONS: Coagulopathy in SAP, a complex phenomenon, is not alleviated by APC treatment. Rather, the patients receiving APC are heading toward normal homeostasis of coagulation slower than patients receiving placebo.


Subject(s)
Anticoagulants/therapeutic use , Pancreatitis/blood , Pancreatitis/drug therapy , Protein C/therapeutic use , Acute Disease , Adult , Anticoagulants/adverse effects , Antithrombin Proteins/analysis , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/etiology , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Pancreatitis/complications , Partial Thromboplastin Time , Protein C/adverse effects , Prothrombin Time
SELECTION OF CITATIONS
SEARCH DETAIL