SUMMARY
Activated protein C (APC) is a plasma serine protease that is capable of antithrombotic, anti-inflammatory, anti-apoptotic, and cell-signaling activities. Animal injury studies show that recombinant APC and some of its mutants are remarkably therapeutic for a wide range of injuries. In particular, for neurologic injuries, APC reduces damage caused by ischemia/reperfusion in the brain, acute brain trauma, and by chronic neurodegenerative conditions. For these neuroprotective effects, APC requires endothelial cell protein C receptor. APC activates cell signaling networks with alterations in gene expression profiles by activating protease activated receptors 1 and 3. To minimize APC-induced bleeding risk, APC variants were engineered to lack > 90 % anticoagulant activity but retain normal cell signaling. The neuroprotective APC mutant, 3K3A-APC which has Lys191-193 mutated to Ala191-193, is very neuroprotective and it is currently in clinical trials for ischemic stroke.
Keywords: Activated Protein C (APC), Stroke, Neuroprotection, Neurogenesis, Traumatic Brain Injury
1. Introduction
The protein C system is a key component of the body’s host-defense systems which evolved to maintain tissue homeostasis and minimize damage from both foreign bodies and from host [1–7]. Initial key findings about the functions of protein C in man came from the discoveries of human hereditary deficiencies; in particular, severe protein C deficiency causes neonatal purpura fulminans which is fatal unless treated aggressively [8, 9]. Moreover, knockout of the murine protein C gene causes pathologic lesions in the brain of embryos and also causes perinatal lethality [10, 11]. In short, results from clinical and murine studies document protein C's essential physiologic roles as an antithrombotic and anti-inflammatory factor.
Clinical data indicate that the brain has particular needs for protein C. Very often, children born with severe protein C deficiency are blind and may also have neurologic deficits [12, 13]. Endogenous APC circulates in an active state at ~ 40 pM in plasma. In the brain, APC is increased during the short ischemic phase of routine carotid endarterectomy in the human brain [14], implying that it might play some neuroprotective role during ischemia. Moreover, circulating APC levels are decreased in stroke patients with antecedent infection/inflammation compared to controls [15]. Finally, it was shown in prospective epidemiologic studies of stroke that plasma protein C levels were inversely associated with the incidence of stroke [16]. Hence, clinical data are consistent with a neuroprotective function for APC. This brief review is focused on the neuroprotective activities of APC [17–22].
2. APC provides neuroprotection for ischemic stroke
APC and its variants have been extensively studied in rodent models of ischemic stroke. Our initial study of murine focal ischemia after middle cerebral artery occlusion (MCAO) (hereafter simplified as murine “stroke”) showed that human APC dose-dependently reduced infarct volume and other markers of brain damage [19]. This success led us to clone murine protein C and then show that recombinant murine wild type (wt)-APC as well as human wt-APC was neuroprotective for murine stroke [20]. Subsequent mechanistic studies showed that wt-APC provided anti-apoptotic activity for ischemic brain microvascular endothelial cells by reducing p53, normalizing the proapoptotic Bax/Bcl-2 ratio, and lowering caspase-3 signaling [21]. Those studies also provided the first in vivo proof of concept evidence for the APC-induced signaling paradigm requiring protease activated receptor (PAR)-1 and endothelial protein C receptor (EPCR) [21].
3. APC is neuronal protective and promotes neurogenesis
Neuronal damage in stroke is greatly reduced by APC. Both in vivo and in vitro studies indicate that APC’s neuronal protection involves signaling and requires PAR-1, PAR-3 and EPCR [18, 22–27]. Current therapy for human stroke patients who meet certain restrictive criteria can involve recombinant tissue plasminogen activator (tPA) therapy which, although beneficial, can cause hemorrhagic transformation; moreover, tPA may possibly be toxic for neurons [28]. Surprisingly, wt-APC, in spite of its potent anticoagulant activity, was vasculoprotective in murine stroke studies and in various test systems it was neuronal-protective both in vivo and in vitro as it reduced tPA-induced hemorrhage and neuronal damage caused by tPA or by NMDA [22, 29, 30]. Thus, we suggested that APC is attractive for adjunctive therapy with tPA. The relationships between APC and neurons became even more fascinating when we discovered that APC promotes neurogenesis in vivo following murine stroke [31] and in vitro in cultures of human embryo-derived neuroprogenitor cells (NPCs) [32]. The ability of 3K3A-APC to promote differentiation and growth of human NPCs requires PAR-1, PAR-3 and sphingosine phosphate receptor-1. Thus, APC and its 3K3A-APC variant are able not only to reduce damage to neurons during and following ischemia reperfusion but also to promote growth and regeneration of neurons [31, 32].
4. APC reduces brain damage following traumatic brain injury
Because wt-APC and 3K3A-APC were found to provide neuroprotection in stroke, they were studied for their ability to reduce damage following traumatic brain injury. For this purpose, a controlled cortical impact injury to mice was used and the effects of APC administration at multiple time points after injury on neuroprotection, angiogenesis and neurogenesis were determined [33, 34]. First, therapy with high doses of 3K3A-APC was safe in the model. Second, APC-treated mice showed improved motor function in a variety of functional tests. Third, APC significantly reduced lesion volumes, increased new blood vessel formation as shown by CD105+/Ki-67+ double immunostaining, and promoted post-traumatic proliferation of neuroblasts in the subventricular zone. Thus, therapy with APC or its variants appears very promising for traumatic brain injury.
5. Cell signaling mechanisms mediate APC’s neuroprotective activities
While the mechanisms for APC’s anticoagulant effects were fairly known last century, the mechanisms for APC’s cell signaling actions are today only partially clear and remain the subject of much ongoing research. In 2001, Joyce et al [35] demonstrated direct effects of APC on endothelial cells and showed that APC was antiapoptotic, anti-inflammatory and altered gene expression profiles. This led others to discover that PAR1 and EPCR were required for APC’s alteration of gene expression [36] and APC’s anti-apoptotic activity [37]. The first in vivo proof of concept for the APC-PAR1/EPCR paradigm for APC-initiated cell signaling came from studies of APC’s neuroprotective actions [21, 22].
Based on many subsequent studies since 2003 of the multiple cytoprotective actions of APC, these activities generally require EPCR and involve APC’s ability to activate PAR1 which can likely involve biased signaling by APC-specific unusual cleavages in PAR1 at Arg46 or in PAR3 [38, 39]. However, various reports for which there is not space here to review also variably implicate other receptors including S1P receptor1, Mac-1 (CD11b/CD18), apolipoprotein E receptor 2, EGFR, Tie-2 and others.
For APC’s neuroprotection and neurogenesis, the following receptors have been identified as contributory, if not strictly required: EPCR, PAR1, PAR3 and S1P receptor 1 [18, 21, 22, 23–27, 30–32]. The finding that signaling-selective 3K3A-APC is normally neuroprotective, like wt-APC, further supports the view that APC’s cytoprotection is due to APC-induced cell signaling [34, 40–43].
6. Signaling-selective 3K3A-APC is in clinical trials for ischemic stroke
To dissect the importance of APC’s anticoagulant activity from its cell signaling activities in cellular and preclinical studies, we and others have engineered APC with selective loss or gain of these two types of APC activities (see [1, 3, 7, 44–48]). For all in vitro and in vivo studies of APC’s neuroprotection, signaling-selective APC was neuroprotective [34, 40–43]. In contrast, an anticoagulant-selective APC mutant lacking normal anti-apoptotic and cytoprotective activities was actually neurotoxic in murine stroke, possibly due to its anticoagulant activity that was 400% of wildtype APC [49]. Thus, clinical trial studies have been based on the principle that APC-initiated cell signaling underlies its neuroprotective actions.
All available studies of the signaling-selective 3K3A-APC, including knowledge for its mechanism of action, enabled moving forward with translation from bench to beside, i.e., undertaking clinical trials of 3K3A-APC that has < 10 % of APC’s anticoagulant activity for ischemic stroke therapy [50, 51]. Preclinical studies of human 3K3A-APC in two animals for toxicity [50] and in two animal stroke models for efficacy [40–43, 52] enabled moving to a phase 1 study [51]. The phase I safety study in normal subjects showed that high dose bolus regimens which resulted in very high transient levels of 3K3A-APC (e.g., plasma levels of 4,500 ng/ml) are safe, including when such high dose boluses are administered every 12 hr over 2.5 days. Moderately severe headache, nausea and vomiting were reported in one of two subjects treated with the maximum dose studied of 0.720 mg/kg, so a 0.540 mg/kg dose was considered the maximum tolerated dose. Elevations in aPTT were minimal, implying bleeding risk is minimal. These very favorable results next need to be confirmed in stroke patients with relevant comorbidities.
The RHAPSODY trial is a Phase 1, dose-escalation and safety trial comparing 3K3A-APC versus placebo in acute stroke patients treated with intravenous recombinant t-PA, intraarterial mechanical thrombectomy [28], or both (https://clinicaltrials.gov/ct2/show/NCT02222714). The trial is being conducted within the NeuroNEXT clinical trials network at 15 sites funded by NINDS. Patients are randomly assigned to treatment group; clinical and imaging endpoints are blinded. A novel hypothesis in RHAPSODY is that the incidence of post-recanalization petechial hemorrhage could be reduced by treatment with 3k3A-APC. The trial is 50% completed as of January 2016.
7. Conclusions
Much has been learned about APC’s neuroprotective activities. There is a remarkable variety of potential indications for APC’s neuroprotective activities, including both acute injury and chronic neurodegenerative conditions [53, 54]. A successful phase 1 clinical study showed that high dose intravenous repeated doses of the signaling-selective APC variant, 3K3A-APC which has < 10 % anticoagulant activity, are safe, setting the stage for ongoing and future clinical trials using this variant for ischemic stroke or for other brain pathologies.
Highlights.
Activated protein C (APC) provides neuroprotection for ischemic stroke
APC is neuronal protective and promotes neurogenesis
APC reduces brain damage following traumatic brain injury
Cell signaling mechanisms mediate APC’s neuroprotective activities
Signaling-selective 3K3A-APC is in clinical trials for ischemic stroke
Acknowledgments
Support from the National Institutes of Health is gratefully acknowledged: HL031950 and HL052246 (J.H.G.); U01 NS088312 (P.D.L.); and NS090904 (B.V.Z.). We thank our collaborators for their contributions to studies of APC’s neuroprotection and we apologize to colleagues whose excellent papers on APC could not be cited due to limited space.
Footnotes
Conflict of interest statement
John H. Griffin is a consultant for ZZ Biotech LLC and inventor for patents involving 3K3A-APC. Berislav V. Zlokovic is a founder and the Chief Scientific Officer of ZZ Biotech LLC, a biotechnology company with a mission to develop APC and its functional mutants for the treatment of stroke and other neurological disorders.
References
- 1.Griffin JH, Zlokovic BV, Mosnier LO. Activated protein C: biased for translation. Blood. 2015;125:2898–2907. doi: 10.1182/blood-2015-02-355974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.McKelvey K, Jackson CJ, Xue M. Activated protein C: A regulator of human skin epidermal keratinocyte function. World J Biol Chem. 2014;5:169–179. doi: 10.4331/wjbc.v5.i2.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rezaie AR. Protease-activated receptor signalling by coagulation proteases in endothelial cells. Thromb Haemost. 2014;112:876–882. doi: 10.1160/TH14-02-0167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Esmon CT. Protein C anticoagulant system--anti-inflammatory effects. Semin Immunopathol. 2012;34:127–132. doi: 10.1007/s00281-011-0284-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bock F, Shahzad K, Vergnolle N, Isermann B. Activated protein C based therapeutic strategies in chronic diseases. Thromb Haemost. 2014;111:610–617. doi: 10.1160/TH13-11-0967. [DOI] [PubMed] [Google Scholar]
- 6.Griffin JH, Zlokovic BV, Mosnier LO. Protein C anticoagulant and cytoprotective pathways. Int J Hematol. 2012;95:333–345. doi: 10.1007/s12185-012-1059-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mosnier LO, Zlokovic BV, Griffin JH. The cytoprotective protein C pathway. Blood. 2007;109:3161–3172. doi: 10.1182/blood-2006-09-003004. [DOI] [PubMed] [Google Scholar]
- 8.Branson HE, Katz J, Marble R, Griffin JH. Inherited protein C deficiency and coumarin-responsive chronic relapsing purpura fulminans in a newborn infant. Lancet. 1983;2:1165–1168. doi: 10.1016/s0140-6736(83)91216-3. Accession Number: 6139528. [DOI] [PubMed] [Google Scholar]
- 9.Chalmers E, Cooper P, Forman K, Grimley C, Khair K, Minford A, et al. Purpura fulminans: recognition, diagnosis and management. Arch Dis Child. 2011;96:1066–1071. doi: 10.1136/adc.2010.199919. [DOI] [PubMed] [Google Scholar]
- 10.Jalbert LR, Rosen ED, Moons L, Chan JC, Carmeliet P, Collen D, et al. Inactivation of the gene for anticoagulant protein C causes lethal perinatal consumptive coagulopathy in mice. J Clin Invest. 1998;102:1481–1488. doi: 10.1172/JCI3011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lay AJ, Liang Z, Rosen ED, Castellino FJ. Mice with a severe deficiency in protein C display prothrombotic and proinflammatory phenotypes and compromised maternal reproductive capabilities. J Clin Invest. 2005;115:1552–1561. doi: 10.1172/JCI24030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dreyfus M, Masterson M, David M, Rivard GE, Muller FM, Kreuz W, et al. Replacement therapy with a monoclonal antibody purified protein C concentrate in newborns with severe congenital protein C deficiency. Semin Thromb Hemost. 1995;21:371–381. doi: 10.1055/s-2007-1000658. [DOI] [PubMed] [Google Scholar]
- 13.Ohga S, Kang D, Kinjo T, Ochiai M, Doi T, Ishimura M, et al. Paediatric presentation and outcome of congenital protein C deficiency in Japan. Haemophilia. 2013;19:378–384. doi: 10.1111/hae.12097. [DOI] [PubMed] [Google Scholar]
- 14.Macko RF, Killewich LA, Fernandez JA, Cox DK, Gruber A, Griffin JH. Brain-specific protein C activation during carotid artery occlusion in humans. Stroke. 1999;30:542–545. doi: 10.1161/01.str.30.3.542. Accession Number: 10066849. [DOI] [PubMed] [Google Scholar]
- 15.Macko RF, Ameriso SF, Gruber A, Griffin JH, Fernandez JA, Barndt R, et al. Impairments of the protein C system and fibrinolysis in infection-associated stroke. Stroke. 1996;27:2005–2011. doi: 10.1161/01.str.27.11.2005. Accession Number: 8898806. [DOI] [PubMed] [Google Scholar]
- 16.Folsom AR, Rosamond WD, Shahar E, Cooper LS, Aleksic N, Nieto FJ, et al. Prospective study of markers of hemostatic function with risk of ischemic stroke. The Atherosclerosis Risk in Communities (ARIC) Study Investigators. Circulation. 1999;100:736–742. doi: 10.1161/01.cir.100.7.736. Accession Number: 10449696. [DOI] [PubMed] [Google Scholar]
- 17.Griffin JH, Zlokovic B, Fernandez JA. Activated protein C: potential therapy for severe sepsis, thrombosis, and stroke. Semin Hematol. 2002;39:197–205. doi: 10.1053/shem.2002.34093. Accession Number: 12124682. [DOI] [PubMed] [Google Scholar]
- 18.Zlokovic BV, Griffin JH. Cytoprotective protein C pathways and implications for stroke and neurological disorders. Trends Neurosci. 2011;34:198–209. doi: 10.1016/j.tins.2011.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shibata M, Kumar SR, Amar A, Fernandez JA, Hofman F, Griffin JH, et al. Anti-inflammatory, antithrombotic, and neuroprotective effects of activated protein C in a murine model of focal ischemic stroke. Circulation. 2001;103:1799–1805. doi: 10.1161/01.cir.103.13.1799. Accession Number: 11282913. [DOI] [PubMed] [Google Scholar]
- 20.Fernández JA, Xu X, Liu D, Zlokovic BV, Griffin JH. Recombinant murine-activated protein C is neuroprotective in a murine ischemic stroke model. Blood Cells Mol Dis. 2003;30:271–276. doi: 10.1016/s1079-9796(03)00034-2. Accession Number: 12737945. [DOI] [PubMed] [Google Scholar]
- 21.Cheng T, Liu D, Griffin JH, Fernandez JA, Castellino F, Rosen ED, et al. Activated protein C blocks p53-mediated apoptosis in ischemic human brain endothelium and is neuroprotective. Nat Med. 2003;9:338–342. doi: 10.1038/nm826. [DOI] [PubMed] [Google Scholar]
- 22.Guo H, Liu D, Gelbard H, Cheng T, Insalaco R, Fernandez JA, et al. Activated protein C prevents neuronal apoptosis via protease activated receptors 1 and 3. Neuron. 2004;41:563–572. doi: 10.1016/s0896-6273(04)00019-4. Accession Number: 14980205. [DOI] [PubMed] [Google Scholar]
- 23.Gorbacheva L, Davidova O, Sokolova E, Ishiwata S, Pinelis V, Strukova S, et al. Endothelial protein C receptor is expressed in rat cortical and hippocampal neurons and is necessary for protective effect of activated protein C at glutamate excitotoxicity. J Neurochem. 2009;111:967–975. doi: 10.1111/j.1471-4159.2009.06380.x. [DOI] [PubMed] [Google Scholar]
- 24.Gorbacheva L, Pinelis V, Ishiwata S, Strukova S, Reiser G. Activated protein C prevents glutamate-and thrombin-induced activation of nuclear factor-kappaB in cultured hippocampal neurons. Neuroscience. 2010;165:1138–1146. doi: 10.1016/j.neuroscience.2009.11.027. [DOI] [PubMed] [Google Scholar]
- 25.Gorbacheva L, Strukova S, Pinelis V, Ishiwata S, Stricker R, Reiser G. NF-kappaB-dependent and - independent pathways in the protective effects of activated protein C in hippocampal and cortical neurons at excitotoxicity. Neurochem Int. 2013;63:101–111. doi: 10.1016/j.neuint.2013.05.008. [DOI] [PubMed] [Google Scholar]
- 26.Gorbacheva LR, Storozhevykh TP, Pinelis VG, Davydova ON, Ishiwata S, Strukova SM. Activated protein C via PAR1 receptor regulates survival of neurons under conditions of glutamate excitotoxicity. Biochemistry (Mosc) 2008;73:717–724. doi: 10.1134/s0006297908060138. Accession Number: 18620539. [DOI] [PubMed] [Google Scholar]
- 27.Gorbacheva LR, Storozhevykh TP, Pinelis VG, Ishiwata S, Strukova SM. Protease-activated receptor (PAR)1-mediated anti-apoptotic effect of activated protein C on glutamate excitotoxicity in hippocampal neurons. Thromb Haemost. 2007;98:1150–1152. doi: 10.1160/th07-03-0228. Accession Number: 18000627. [DOI] [PubMed] [Google Scholar]
- 28.Amar AP, Griffin JH, Zlokovic BV. Combined neurothrombectomy or thrombolysis with adjunctive delivery of 3K3A-activated protein C in acute ischemic stroke. Front Cell Neurosci. 2015;9:344. doi: 10.3389/fncel.2015.00344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Liu D, Cheng T, Guo H, Fernandez JA, Griffin JH, Song X, et al. Tissue plasminogen activator neurovascular toxicity is controlled by activated protein C. Nat Med. 2004;10:1379–1383. doi: 10.1038/nm1122. [DOI] [PubMed] [Google Scholar]
- 30.Cheng T, Petraglia AL, Li Z, Thiyagarajan M, Zhong Z, Wu Z, et al. Activated protein C inhibits tissue plasminogen activator-induced brain hemorrhage. Nat Med. 2006;12:1278–1285. doi: 10.1038/nm1498. [DOI] [PubMed] [Google Scholar]
- 31.Thiyagarajan M, Fernandez JA, Lane SM, Griffin JH, Zlokovic BV. Activated protein C promotes neovascularization and neurogenesis in postischemic brain via protease-activated receptor 1. J Neurosci. 2008;28:12788–12797. doi: 10.1523/JNEUROSCI.3485-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Guo H, Zhao Z, Yang Q, Wang M, Bell RD, Wang S, et al. An activated protein C analog stimulates neuronal production by human neural progenitor cells via a PAR1-PAR3-S1PR1-Akt pathway. J Neurosci. 2013;33:6181–6190. doi: 10.1523/JNEUROSCI.4491-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Petraglia AL, Marky AH, Walker C, Thiyagarajan M, Zlokovic BV. Activated protein C is neuroprotective and mediates new blood vessel formation and neurogenesis after controlled cortical impact. Neurosurgery. 2010;66:165–171. doi: 10.1227/01.NEU.0000363148.49779.68. discussion 71-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Walker CT, Marky AH, Petraglia AL, Ali T, Chow N, Zlokovic BV. Activated protein C analog with reduced anticoagulant activity improves functional recovery and reduces bleeding risk following controlled cortical impact. Brain Res. 2010;1347:125–131. doi: 10.1016/j.brainres.2010.05.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Joyce DE, Gelbert L, Ciaccia A, DeHoff B, Grinnell BW. Gene expression profile of antithrombotic protein c defines new mechanisms modulating inflammation and apoptosis. J Biol Chem. 2001 Apr 6;276(14):11199–11203. doi: 10.1074/jbc.C100017200. [DOI] [PubMed] [Google Scholar]
- 36.Riewald M, Petrovan RJ, Donner A, Mueller BM, Ruf W. Activation of endothelial cell protease activated receptor 1 by the protein C pathway. Science. 2002;296:1880–1882. doi: 10.1126/science.1071699. [DOI] [PubMed] [Google Scholar]
- 37.Mosnier LO, Griffin JH. Inhibition of staurosporine-induced apoptosis of endothelial cells by activated protein C requires protease-activated receptor-1 and endothelial cell protein C receptor. Biochem J. 2003 Jul 1;373(Pt 1):65–70. doi: 10.1042/BJ20030341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mosnier LO, Sinha RK, Burnier L, Bouwens EA, Griffin JH. Biased agonism of protease-activated receptor 1 by activated protein C caused by noncanonical cleavage at Arg46. Blood. 2012;120:5237–5246. doi: 10.1182/blood-2012-08-452169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Burnier L, Mosnier LO. Novel mechanisms for activated protein C cytoprotective activities involving noncanonical activation of protease-activated receptor 3. Blood. 2013;122:807–816. doi: 10.1182/blood-2013-03-488957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang Y, Thiyagarajan M, Chow N, Singh I, Guo H, Davis TP, et al. Differential neuroprotection and risk for bleeding from activated protein C with varying degrees of anticoagulant activity. Stroke. 2009;40:1864–1869. doi: 10.1161/STROKEAHA.108.536680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang Y, Zhang Z, Chow N, Davis TP, Griffin JH, Chopp M, et al. An activated protein C analog with reduced anticoagulant activity extends the therapeutic window of tissue plasminogen activator for ischemic stroke in rodents. Stroke. 2012;43:2444–2449. doi: 10.1161/STROKEAHA.112.658997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang Y, Zhao Z, Chow N, Rajput PS, Griffin JH, Lyden PD, et al. Activated protein C analog protects from ischemic stroke and extends the therapeutic window of tissue-type plasminogen activator in aged female mice and hypertensive rats. Stroke. 2013;44:3529–3536. doi: 10.1161/STROKEAHA.113.003350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Guo H, Singh I, Wang Y, Deane R, Barrett T, Fernandez JA, et al. Neuroprotective activities of activated protein C mutant with reduced anticoagulant activity. Eur J Neurosci. 2009;29:1119–1130. doi: 10.1111/j.1460-9568.2009.06664.x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 44.Mosnier LO, Gale AJ, Yegneswaran S, Griffin JH. Activated protein C variants with normal cytoprotective but reduced anticoagulant activity. Blood. 2004;104:1740–1744. doi: 10.1182/blood-2004-01-0110. [DOI] [PubMed] [Google Scholar]
- 45.Mosnier LO, Yang XV, Griffin JH. Activated protein C mutant with minimal anticoagulant activity, normal cytoprotective activity, and preservation of thrombin activable fibrinolysis inhibitor-dependent cytoprotective functions. J Biol Chem. 2007;282:33022–33033. doi: 10.1074/jbc.M705824200. [DOI] [PubMed] [Google Scholar]
- 46.Mosnier LO, Zlokovic BV, Griffin JH. Cytoprotective-selective activated protein C therapy for ischaemic stroke. Thromb Haemost. 2014;112:883–892. doi: 10.1160/TH14-05-0448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wildhagen KC, Lutgens E, Loubele ST, ten Cate H, Nicolaes GA. The structure-function relationship of activated protein C. Lessons from natural and engineered mutations. Thromb Haemost. 2011;106:1034–1045. doi: 10.1160/TH11-08-0522. [DOI] [PubMed] [Google Scholar]
- 48.Quinn LM, Drakeford C, O'Donnell JS, Preston RJ. Engineering activated protein C to maximize therapeutic efficacy. Biochem Soc Trans. 2015 Aug 1;43(4):691–695. doi: 10.1042/BST20140312. [DOI] [PubMed] [Google Scholar]
- 49.Wang Y, Sinha RK, Mosnier LO, Griffin JH, Zlokovic BV. Neurotoxicity of the anticoagulant-selective E149A-activated protein C variant after focal ischemic stroke in mice. Blood Cells Mol Dis. 2013;51:104–108. doi: 10.1016/j.bcmd.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Williams PD, Zlokovic BV, Griffin JH, Pryor KE, Davis TP. Preclinical safety and pharmacokinetic profile of 3K3A-APC, a novel, modified activated protein C for ischemic stroke. Curr Pharm Des. 2012;18:4215–4222. doi: 10.2174/138161212802430413. Accession Number: 22632606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lyden P, Levy H, Weymer S, Pryor K, Kramer W, Griffin JH, et al. Phase 1 safety, tolerability and pharmacokinetics of 3K3A-APC in healthy adult volunteers. Curr Pharm Des. 2013;19:7479–7485. doi: 10.2174/1381612819666131230131454. Accession Number: 24372304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wang Y, Zhao Z, Chow N, Ali T, Griffin JH, Zlokovic BV. Activated protein C analog promotes neurogenesis and improves neurological outcome after focal ischemic stroke in mice via protease activated receptor 1. Brain Res. 2013;1507:97–104. doi: 10.1016/j.brainres.2013.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Han MH, Hwang SI, Roy DB, Lundgren DH, Price JV, Ousman SS, et al. Proteomic analysis of active multiple sclerosis lesions reveals therapeutic targets. Nature. 2008 Feb 28;451(7182):1076–1081. doi: 10.1038/nature06559. [DOI] [PubMed] [Google Scholar]
- 54.Zhong Z, Ilieva H, Hallagan L, Bell R, Singh I, Paquette N, et al. Activated protein C therapy slows ALS-like disease in mice by transcriptionally inhibiting SOD1 in motor neurons and microglia cells. J Clin Invest. 2009;119:3437–3449. doi: 10.1172/JCI38476. [DOI] [PMC free article] [PubMed] [Google Scholar]