Next Article in Journal
Plant-Based Biosynthesis of Copper/Copper Oxide Nanoparticles: An Update on Their Applications in Biomedicine, Mechanisms, and Toxicity
Next Article in Special Issue
Role of Purinergic Signalling in Endothelial Dysfunction and Thrombo-Inflammation in Ischaemic Stroke and Cerebral Small Vessel Disease
Previous Article in Journal
Xanthurenic Acid Is the Main Pigment of Trichonephila clavata Gold Dragline Silk
Previous Article in Special Issue
Monitoring and Modulating Inflammation-Associated Alterations in Synaptic Plasticity: Role of Brain Stimulation and the Blood–Brain Interface
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Thrombin in Central Nervous System Injury and Disease

by
Nathan A. Shlobin
1,*,
Meirav Har-Even
2,3,*,
Ze’ev Itsekson-Hayosh
4,5,
Sagi Harnof
6 and
Chaim G. Pick
2,3,7,8,*
1
Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
2
Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
3
Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
4
Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
5
Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer 5262000, Israel
6
Department of Neurosurgery, Beilinson Hospital, Rabin Medical Center, Tel Aviv University, Petah Tikva 4941492, Israel
7
Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
8
Center for Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 6997801, Israel
*
Authors to whom correspondence should be addressed.
Biomolecules 2021, 11(4), 562; https://doi.org/10.3390/biom11040562
Submission received: 27 February 2021 / Revised: 4 April 2021 / Accepted: 7 April 2021 / Published: 12 April 2021
(This article belongs to the Special Issue Novel Biomolecules in Neuro-ThromboInflammation)

Abstract

:
Thrombin is a Na+-activated allosteric serine protease of the chymotrypsin family involved in coagulation, inflammation, cell protection, and apoptosis. Increasingly, the role of thrombin in the brain has been explored. Low concentrations of thrombin are neuroprotective, while high concentrations exert pathological effects. However, greater attention regarding the involvement of thrombin in normal and pathological processes in the central nervous system is warranted. In this review, we explore the mechanisms of thrombin action, localization, and functions in the central nervous system and describe the involvement of thrombin in stroke and intracerebral hemorrhage, neurodegenerative diseases, epilepsy, traumatic brain injury, and primary central nervous system tumors. We aim to comprehensively characterize the role of thrombin in neurological disease and injury.

1. Introduction

Thrombin is a Na+-activated allosteric serine protease of the chymotrypsin family central to blood coagulation with further involvement in inflammatory responses, cell protection, and apoptosis [1,2,3,4,5,6]. Thrombin is a large, spherical molecule, with a groove, consisting of a light A-chain and heavy B-chain with two anion-binding exosites [7,8,9]. The structure of thrombin allows binding of substrates to promote signaling through a myriad of different pathways [7,10,11]. Exosite I recognizes fibrinogen, fibrin, factor Va, thrombomodulin, and hirudin, while exosite II recognizes glycosaminoglycans, including heparin sulfate and glycoprotein Ibα [8,9]. Prothrombin, the inactive precursor of thrombin, is generated within the liver for circulation in the plasma until conversion into active thrombin in the coagulation cascade [8,12]. The serine protease inhibitor antithrombin III is responsible for irreversible inactivation of thrombin via interaction with heparan sulfates, while exogenous compounds including hirudin and heparin may inhibit thrombin by binding to exosites [8,13,14,15,16]. Thrombin is involved in wound healing, arterial and venous thrombosis, and the pathogenesis of conditions, including atherosclerosis, sepsis, and cancer [17]. It is clear that thrombin is involved in neurological disease [8,18]. In this review, we explore the mechanisms of thrombin action, localization, and functions in the central nervous system (CNS) and describe the involvement of thrombin in stroke and intracerebral hemorrhage, neurodegenerative diseases, epilepsy, traumatic brain injury, and primary CNS tumors. We aim to comprehensively characterize the role of thrombin in neurological disease and injury (Figure 1).

2. Mechanisms of Thrombin Action

2.1. Coagulation

In the coagulation cascade, tissue factor complexes with factor VIIIa, generating factors IXa and Xa [1,2,3,17,19]. Small quantities of Xa generate miniscule quantities of thrombin, activating factor XI, and cofactors VIII and V [1,2,3,17,19]. The VIII–IXa complex generates sufficient Xa to promote the formation of the prothrombinase complex, consisting of factors Va and Xa, Ca2+, and anionic phospholipids [1,2,3,17,19]. Within the prothrombinase complex, factor Xa proteolytically cleaves prothrombin to meizothrombin, with further cleavage to thrombin [1,2,3,17,19]. Paradoxically, thrombin exerts both procoagulant and anticoagulant effects [1,2,3,17,19]. In the procoagulant pathway, thrombin converts fibrinogen to an insoluble fibrin clot to which platelets adhere in order to begin wound repair [1,2,3,17,19]. Activation of transglutaminase factor XIII to stabilize the fibrin clot, TAF1 inhibition of fibrinolysis, and proteolytic action of factors V, VIII, and XI reinforce the procoagulant actions [1]. In the anticoagulant pathway, thrombin interacts with protease-activated receptors (PAR), a family of G-protein coupled receptors activated through cleavage of a portion of an extracellular domain. Binding of thrombin to thrombomodulin augments the activation of protein C on the endothelial protein C receptor (ECPR) while restricting cleavage of fibrinogen and PAR1 [1,2,3,17,19]. Protein C cleaves and inactivates factors Va and VIII to prevent further thrombin generation, while the serine protease inhibitor antithrombin irreversibly inhibits thrombin along with heparin [1,2,3,17,19]. These divergent actions promote a balance between the procoagulant and anticoagulant pathways [1,2,3,17,19].

2.2. Thrombotic and Immune Functions

Pleiotropic effects of thrombin have been described primarily in in vitro studies [17]. Thrombin affects the activity of platelets, fibroblasts, vascular smooth muscle cells, endothelial cells, monocytes, and T lymphocytes [20,21,22,23,24]. Thrombin exerts mitogenic activity on vascular smooth muscle cells and endothelial cells via PARs and modulates vascular permeability, vascular tone, and angiogenesis [17,25,26,27,28,29,30,31,32,33]. Mitogenic effects result from the action of basic fibroblast growth factor and sequestering of thrombin in subendothelial basement membranes [29,30]. Changes in vascular tone are secondary to PAR1-initiated endothelial-dependent relaxation mediated by nitric oxide and polarizing factors [33]. Increases in vascular permeability occur due to changes in the distribution of vascular endothelial-cadherin and associated catenins and actin–myosin interactions [31,32]. This results in plasma protein leakage, creating a proangiogenic matrix [34,35,36]. Decreased cyclic adenosine monophosphate-mediated attachment of endothelial cells promotes angiogenesis coupled with potentiation of vascular endothelial growth factor (VEGF)-induced endothelial cell regulation, increased transcription of VEGF via the production of reactive oxygen species (ROS) and expression of hypoxia-inducible factor 1, upregulation of VEGF receptor expression, and increased levels of αV/β3 integrin [37,38,39,40,41]. Thrombin promotes inflammation and intimal hyperplasia via dysfunctional mixed phenotype intermediates of vascular progenitors and the activation of monocytes, T lymphocytes, mast cells, and endothelial cells [42,43]. Thrombin also affects processes involved in tissue repair, including PAR-1 mediated induction of cytokines promoting angiogenesis, leukocyte migration, and edema formation [34,44,45].

2.3. Cellular Protection and Apoptosis

Thrombin is involved in cell protection and induces apoptosis through the activation of PAR1 via the guanosine triphosphate binding protein RhoA and the protein kinase C pathway [11,46,47]. PAR1 is activated via cleavage by thrombin into tethered activation peptide ligands that enable transmembrane signaling [48]. After cleavage, some PAR1 receptors are internalized, while approximately 20–60% remain on the cell surface [49]. The activity of thrombin depends on thrombin concentration and length of exposure of thrombin to the cellular environment (Figure 2). PAR1 receptors are cleaved more rapidly at high thrombin concentrations, enabling a greater cellular response [49]. Short exposure to low thrombin concentrations activates pathways for cellular protection, while prolonged exposure to thrombin stimulates apoptosis [4]. Given the ability of PARs to form homodimers and heterodimers, the specific dimer also determines which signal transduction cascade is active [50,51]. Interaction of the activated protein C (APC) with endothelial protein C receptor (EPCR) promotes anti-inflammatory effects, cellular protection, and endothelial barrier stabilization by switching PAR1 signal transduction to cytoprotective and regenerative functions [46,52,53,54]. EPCR is also involved in endocytosis via interaction with the lipid raft plasma membrane protein caveolin-1 [4]. Binding of the APC or protein C to EPCR prompts activation of PAR1 after dissociation of EPCR from caveolin 1 [4]. At low thrombin concentrations, PAR1 activation is mediated by the APC–EPCR, promoting cytoprotective Gαi signaling and subsequent Rac1 activation. High thrombin concentrations promote PAR1 activation via Gαq and Gα12/13 signaling and subsequent RhoA activation and eliminate cytoprotective effects of the APC-EPCR via activation of PAR4 [4,46,52,54,55,56]. The combined effect is a switch to thrombin-mediated cellular degeneration [46]. Additionally, scaffold proteins β-arrestin 1 and 2 are involved in signaling dependent on and independent of G-protein-coupled receptors [4]. A dose-dependent switch mediates the transition to G-protein-independent signaling, inducing cytoskeletal organization [57,58,59]. However, activation of cellular pathways via a receptor depends on ligand bias, receptor bias, and cell bias, indicative of specific conditions that promote the activation of distinct pathways that have yet to be determined [4,60].

3. Thrombin in the Central Nervous System

3.1. Localization in Brain

Prothrombin and thrombin have been localized to neurons and glial cells in the CNS [61]. Similarly, factor X and inhibitors of thrombin activity, including antithrombin III and the protease nexin-1 (PN-1), are locally expressed in the brain, with much of the PN-1 localized around blood vessels [62,63,64,65]. Prothrombin mRNA expression is greatest in the cerebral cortex and moderate in the hippocampus and cerebellum [66]. The hippocampus displays greater labeling of thrombin in the pyramidal layers and lower labeling in fiber layers on immunohistochemistry [67]. Prothrombin expression and thrombin activity in the nervous system are highly regulated in physiological and pathological states [68,69]. Similarly, PARs are expressed in neurons, oligodendrocytes, microglia, and astrocytes [61,70,71,72], indicating that thrombin may be active in these cells under physiological conditions. Similarly, PAR1 is found in the pyramidal layers of the hippocampus, and PAR3 and PAR4 are found in all cortical layers and the thalamus of rat brains [71]. Rat neurons and astrocytes contain all PAR receptors [73,74]. While thrombin utilizes all four PARs to engage signal transduction pathways, PAR1 activation is primarily responsible for the actions of thrombin in the CNS [8]. High concentrations of thrombin increase brain damage, while low thrombin concentrations and thrombin preconditioning are neuroprotective [61,75].

3.2. Neuro-Physiological Functions

Thrombin has myriad physiological functions in the CNS. First, thrombin plays a role in neuronal development, given the colocalization and similar developmental pattern of prothrombin mRNA and PAR1 in the rat brain [76,77]. Second, thrombin exerts mitogenic functions to enable cellular proliferation and differentiation—as described above. Mitogen-activated protein kinases (MAPKs) regulate cellular proliferation and differentiation, while PAR1 mediates the mitogenic action in astrocytes and microglia [73,78,79,80]. Nerve growth factor and endothelin-1 synthesis enable astrocyte proliferation [81,82,83]. Additional signal transduction cascades specific to cell type involve the PI 3-kinase pathway and phospholipase C (PLC)/Ca2+/protein kinase C (PKC) pathway [84]. Third, thrombin signaling alters cell morphology and enables cell migration. Thrombin alters the morphology of astrocytes, fetal neurons, and neuroblastoma cells via neurite outgrowth, stellate retraction, and cytoskeleton rearrangement to enable growth cone guidance and cell migration [81,82,83,85,86]. Fourth, thrombin regulates synaptic transmission. This has been demonstrated particularly in the CA3 layer of the hippocampus but not the CA1 layer [87]. Fifth, thrombin regulates synaptic plasticity [87]. Thrombin affects long term potentiation in the hippocampus through a series of pathways mediated by PAR1 [88,89,90,91,92,93,94,95]. High thrombin concentrations prompt a slow onset long-term potentiation dependent on the N-methyl-D-aspartate (NMDA) receptor [88,89]. Low thrombin concentrations promote long-term potentiation dependent on voltage-gated calcium channels and mGluR-5 through APC [88,89].

3.3. Neuro-Pathological Functions

Thrombin also plays a role in pathological processes in the CNS. Thrombin action is greatest under conditions leading to disruption of the blood–brain barrier (BBB) [96]. Thrombin induces damage to the BBB and an associated increase in permeability through a variety of mechanisms. Thrombin leads to F-actin fiber increase, destruction of tight junction, nitric oxide release, and PAR-1 dependent production of mitochondrial and cytosolic ROS [97]. Disruption of the BBB also results from thrombin-related expression of matrix metalloproteinases (MMPs) in brain tissue, particularly in pericytes and in activated microglia [98,99,100]. Similarly, thrombin is involved in brain inflammation. Thrombin activates microglia through p38 MAPK and c-Jun N-terminal kinase (JNK) in processes mediated by PAR1, PAR3, and PAR4 and has been associated with glial scar formation [73,74,79,101]. Thrombin also activates astrocytes through pro-inflammatory compounds, including arachidonic acid, nitric oxide, the chemokine growth-regulated oncogene/cytokine-induced neutrophil chemoattractant-1, and interleukin (IL)-8 [102,103]. Thrombin increase enhances the expression and release of pro-inflammatory factors, such as CD-40, cyclooxygenase 2, inducible nitric oxide synthase, tumor necrosis factor-α, IL-1α/β, IL-6, and IL-12 [79]. These cytokines may exert positive feedback on thrombin production [104]. Additionally, thrombin promotes neurotoxicity. Thrombin induces shrinkage of striatal tissue dependent on activation of microglia, extracellular signal-related kinase, and MAPK-related pathways [105,106]. Concomitant exposure to MMP-9 increases neurotoxicity as PAR1 converts pro-MMP-9 to active MMP-9 [107,108]. Thrombin also cleaves apolipoprotein E (apoE) to a fragment with a cytotoxic domain [109,110]. Conversely, injured brain cells release neurotoxic thrombin [111]. Moreover, thrombin promotes maladaptive synaptic plasticity that may result in seizures [89,112]. Furthermore, thrombin is involved in intracerebral coagulation, as in the remainder of the body [113]. These pathological functions, mediated by thrombin in neurological conditions and injuries, will be further elaborated.

4. Neurodegenerative Diseases

4.1. Alzheimer’s Disease

The role of thrombin in Alzheimer’s disease (AD) has been well characterized. Thrombin accumulates in senile plaques, amyloid deposits, neurofibrillary tangles, and microvessels in the brains of AD patients, while prothrombin mRNA is expressed in neurons and glial cells [114,115]. In a rat model, the modulation of PAR (particularly PAR1) expression was demonstrated in hippocampal astrocytes and microglia [116]. AD also promotes the release of thrombin through β-amyloid (Aβ) activation of factor XII and release of the transcription factor hypoxia inducible factor 1α (HIF-1α) [117,118,119]. Thrombin represents a convergence point for AD risk factors [120]. In vitro experiments have demonstrated intracellular aggregates of the microtubule-associated tau protein in hippocampal neurons arising from the ability of thrombin to proteolyze tau protein but inability to process phosphorylated tau protein [121]. Neurotoxic tau deposits leading to apoptosis of hippocampal neurons result from thrombin-mediated hyperphosphorylation and accumulation of tau via the of PAR1/4 and the extracellular signal-regulated kinase (ERK)1/2 pathway [121,122]. Thrombin also cleaves β-amyloid precursor protein, promoting Aβ accumulation [123,124]. Intracellular Ca2+ influx and oxidative stress compound Aβ neurotoxicity [125]. Although PN-1 protects neurons against neurotoxicity resulting from Aβ accumulation, thrombin–PN-1 complex formation attenuates the activity of PN-1 [126,127]. Thrombin promotes microglial NAPDH oxidase production of ROS and expression of pro-inflammatory IL-8 and integrins, two key pathophysiological processes in AD, leading to further cell death [8,118,128]. Intracerebral administration of thrombin in rats increases apoE levels, leading to Aβ accumulation and ensuing cognitive difficulties [119]. The direct thrombin inhibitor dabigatran reduces expression of inflammatory cells, markers of oxidative stress including ROS, and tau pathology in vivo [129,130]. Accordingly, investigators have suggested the use of direct thrombin inhibitors in the treatment of AD due to their selectivity in inhibiting thrombin and relatively mild side effect profile [131]. However, whether thrombin prompts neurodegeneration and leads to formation of amyloid plaques and neurofibrillary tangles in vivo remains to be seen.

4.2. Parkinson’s Disease

Thrombin is associated with modulation of neurological injury and progression of Parkinson’s disease (PD). Prothrombin and PAR1 are upregulated in astrocytes positive for glial fibrillary acid protein in the brains of PD patients, and expression of PARs is increased in the vessel wall of the substantia nigra pars compacta [8]. Loss of dopaminergic cells, neuroinflammation, and oxidative stress occur after thrombin injection into the substantia nigra [132,133,134,135,136]. Expression of pro-apoptotic proteins caspase-3 and p53 is upregulated in dopaminergic neurons of the substantia nigra, while pro-inflammatory molecules including nitric oxide, IL-1α/β, IL-6 and TNF-α are expressed due to microglial activation by thrombin [8,132,134,136]. The timing and dose of thrombin administration is important in understanding the pathophysiological role of thrombin in PD. Delayed preconditioning with thrombin protects against cell damage resulting from infusion of 6-hydroxydopamine [137]. Coadministration of 6-hydroxydopamine and thrombin or PAR1 agonists amplifies neuronal damage and behavioral deficits, while previous treatment with PAR1 antagonists eliminates the neuroprotective effects of preconditioning [137]. However, thrombin neurotoxicity may also be independent of the PAR1 pathway, and PAR1 activation may be neuroprotective given the absence of PAR1 in microglia in the substantia nigra pars compacta [8,138]. Upregulation of PAR1 in astrocytes may have restorative action on dopaminergic neurons, preventing their degeneration and cell death in PD [139]. Similarly, thrombin preconditioning reduces dopaminergic terminal loss and ventricular enlargement, while PAR1 antagonists exacerbate neurological deficits resulting from 6-hydroxydopamine administration [140,141]. Interestingly, in the substantia nigra pars compacta of patients with PD, neurons, microglia, and oligodendrocytes lack PARs [139]. However, PAR4 is involved in dopaminergic neuron loss in the substantia nigra upon striatal thrombin injection [71], indicating PAR4 may modulate thrombin-related pathogenesis in PD. As with AD, dabigatran, a selective thrombin inhibitor widely prescribed as an oral anticoagulant, has been utilized in animal models of PD. Dabigatran suppresses thrombin accumulation in the substantia nigra, decreasing the expression of pro-inflammatory cytokines and reducing oxidative stress [142,143]. Lipopolysaccharide-binding protein has also been shown to reverse the amyloid accumulation of fibrin induced by bacterial lipopolysaccharide and occurring in the blood of patients with PD [144,145]. Additional studies are necessary to characterize the utility of these treatments.

4.3. Multiple Sclerosis

The role of thrombin in multiple sclerosis (MS) is less clear than in AD or PD. Progressive axonal loss in animal models of MS is associated with fibrin accumulation in cerebral vasculature [146]. Thrombin activity is associated with BBB disruption, microglial activation, inflammatory demyelination, and axonal damage, indicating that thrombin may be involved in the pathogenesis of MS [147]. Mouse models of experimental autoimmune encephalitis (EAE), the most commonly used experimental model for MS, demonstrate a large increase in thrombin activity prior to appearance of motor impairment [147,148,149]. Thrombin activity continues to increase until the peak of clinical disease [147,148,149]. Thrombin activity begins in the early stage of the disease preceding demyelination and is associated with the progression of disease [147]. Thrombin activity and fibrin deposition are closely associated with increased microglial activation and the extent of demyelination [147]. Studies examining human patients have not reported increased thrombin in the CNS [150]. However, PAR1 mRNA expression and surface density in platelets and megakaryocytes and plasma factor XII activity are greater in patients with MS [151,152,153]. Similarly, oligodendrocytes express PAR1 receptors, while PAR2 activation secondary to the action of macrophages promotes oligodendrocyte death in the EAE model [154,155]. Additionally, EAE animals exhibit increased BBB permeability early in the clinical course [156]. Given that PAR1 inhibitors preserve the BBB and reduce demyelination and inflammatory infiltration of the CNS while decreasing thrombin levels in an EAE mouse model, PAR1 inhibitors may play a role in treatment of MS [157]. Early PN-1 changes may also be a target for thrombin-modulating drugs in MS [149]. Temporal pharmacological enhancement of endogenous APC generation via a selective recombinant protein C activator thrombin analog may represent another treatment option [158]. However, future studies are necessary to completely characterize the role of thrombin in the neuroinflammation present in MS.

5. Intracerebral Hemorrhage and Stroke

Thrombin is involved in stroke and intracranial hemorrhage (ICH). Elevated thrombin increases the risk of acute ischemic stroke [159]. Conversely, ischemia leads to increased levels of thrombin and prothrombin via activation of factor Xa, and increased BBB permeability permits thrombin influx from the bloodstream [8,160,161]. Thrombin activity is associated with infarct volume [162]. Similarly, thrombin activity eluted from clots originating from patients with atrial fibrillation differs from that of patients with atherosclerosis, indicating its potential role as a diagnostic marker [163]. Thrombin exerts dose-dependent effects. Low concentrations protect neurons and astrocytes in the hippocampus against oxygen glucose deprivation, hypoglycemia, and ROS, while higher concentrations promote hippocampal and motor neuron cell death due to unknown mechanisms of PAR1 neurotoxicity [8,164,165,166]. Focal ischemia and ICH can cause intracerebral edema associated with an increase in thrombin [113,167,168,169]. Thrombin determines the release of pro-inflammatory factors, including iron and MMPs [79,136,168,170]. Iron increases the risk of brain edema after ICH, as transferrin containing iron overwhelms neuroprotective effects at low thrombin concentrations [171,172]. MMPs augment damage to neural tissue [107]. Separately, expression of pro-apoptotic proteins such as Bim due to increased cyclin-dependent kinase 4 stimulation in hypoxic conditions facilitates apoptosis [173,174]. PAR1 is primarily responsible for mediation of thrombin activity, despite complex regulation of PAR1, PAR3, and PAR4 [8]. PAR1 activation promotes disruption of the extracellular matrix and increased permeability of the BBB in addition to neuronal damage and increased size of infarcts [72,175]. NMDA receptor stimulation via increased astrocyte glutamate release secondary to PAR1 activation may also lead to excitotoxicity and subsequent BBB disruption [91,176,177]. The formation of glial scars via PAR1 activation after stroke or ICH decreases the potential for regeneration of neural tissue [101,178]. Thrombin preconditioning reduces the size of the infarct in ischemic stroke and edema in ICH [68,179,180]. PAR1 antagonists eliminate thrombin preconditioning-mediated neuroprotection [75]. The effect of hirudin is equivocal, as authors have described reversal of neuroprotective thrombin preconditioning, while others have described reduced infarct volumes upon administration following occlusion of the middle cerebral artery [181,182]. Thrombin preconditioning may include ceruloplasmin upregulation to promote tolerance to edema, and JNK may promote neuroprotection [183,184]. The balance between thrombin and PN-1 may augment repair processes in stroke, given unchanged expression levels under ischemic conditions [185,186]. Although dabigatran is utilized in stroke prophylaxis in patients with atrial fibrillation, its efficacy relative to other treatments such as rivaroxaban is equivocal [187,188,189]. Determining the optimal dosing of dabigatran and utility with concomitant antiplatelet therapy may be particularly important [190]. Future studies should continue to identify mechanisms of thrombin involvement in stroke and ICH.

6. CNS Infections

Thrombin has been implicated in infections with neurological symptomatology. Translocation of the BBB is an important step in the pathogenesis of CNS infections [191,192]. Increased permeability of the BBB due to thrombin increases the ability of pathogens to cross the blood–brain barrier. Meningitis-causing microorganisms enter the brain through transcellular penetration of the BBB [191]. Thrombin-activated fibrinolysis inhibitor (TAFI) likely mediates inhibition of the complement system and activation of systemic complications and inflammation in pneumococcal meningitis [193]. Similarly, TAFI genotype is associated with the risk of meningococcal disease and death [194]. Activation of TAFI mediates disseminated intravascular coagulation and sepsis in patients with meningococcal sepsis [195]. Expression of endothelial thrombomodulin and endothelial protein C receptor is lower in patients with meningococcal sepsis, emphasizing the role of dysfunction of the thrombin-mediated anticoagulation pathways in severe disease [196]. Additionally, the role of thrombin in CNS infections is best characterized in the pathogenesis of human immunodeficiency virus (HIV). Thrombin generation rises by 24–48% while anticoagulants including antithrombin and protein C decrease in computational models of untreated HIV infection [197]. A concentration-dependent mechanism may underlie acceleration of the HIV-induced cell-fusion rate by thrombin [8]. HIV promotes disruption of the BBB, perhaps promoting an influx of prothrombin or thrombin, despite decreased thrombin generation in the plasma [198,199]. HIV also increases coagulation potential, augmenting the risk of ischemic cerebral infarction and intracranial venous thromboembolism and atherosclerotic disease [197]. Thrombin may be involved in the CNS pathogenesis of HIV via stimulation of T cell motility and production of pro-inflammatory cytokines to moderate the cross-talk between the coagulation cascade and adaptive immune system in areas of vascular injury [200]. Recent evidence suggests that thrombin plays a role in the CNS sequelae of HIV. HIV-associated encephalitis and dementia involve neuroinflammation and continual neuronal damage [8]. HIV-dementia involves changes mediated by PARs, including altered morphology, neurite retraction, or neurotoxicity [8]. PAR2 is upregulated in neurons, suggesting involvement of thrombin in HIV-associated dementia [201]. Similarly, patients with HIV-associated encephalitis exhibit increased levels of prothrombin mRNA and protein and upregulated PAR1 in astrocytes [176]. Neuroinflammation in HIV patients may be PAR1-dependent [202]. The direct relationship between thrombin and other neuroinfectious diseases continues to be investigated. Schizont-stage Plasmodium falciparum induces low levels of primary human microvascular endothelial cell death and prolongs thrombin-induced barrier disruption [203]. β-arrestin 2, a downstream component of thrombin, protects against neurological dysfunction in herpes simplex virus-1 induced encephalitis [204]. Varicella zoster virus encephalitis may promote a vasculopathy [205]. Additional research is necessary to precisely characterize potential additional mechanisms through which thrombin promotes CNS infections.

7. Mild Traumatic Brain Injury (mTBI)

The involvement of thrombin in mild traumatic brain injury (mTBI) has begun to be investigated. Thrombin levels rise until one-hour post-trauma and increase again after 72 h with a concomitant increase in PAR1 at both time points, indicative of astrocyte activation [206]. Similarly, traumatic brain injury is associated with disruption of the BBB, allowing thrombin to enter and promote neurotoxicity [68]. In cases of trauma-induced amnesia, recovery from amnesia occurs when thrombin activity normalizes in the hippocampus [207]. Rescue from trauma-induced amnesia can be accomplished by inhibiting thrombin activity or blocking PAR1 [208]. A recent study determined that downregulation of hippocampal astrocyte glutamate transporters by thrombin following TBI was associated with depression in mice [209]. Inhibition of PAR1 or Rho kinase decreased depressive symptoms [209]. Further investigation regarding the role of thrombin in the pathogenesis of TBI is warranted.

8. Epilepsy

The role of thrombin in epilepsy is being increasingly explored. Thrombin increases sensitivity to seizure-like activity [206]. Seizures are commonly associated with conditions that compromise BBB function, including stroke and intracerebral hemorrhage, TBI, and CNS infections through regional destruction of the BBB [210,211,212]. Increased permeability of the BBB stimulates seizures due to increased exposure of the brain to thrombin and other serum components [213]. BBB breakdown activates the coagulation cascade irrespective of the presence of intracerebral hemorrhage, generating additional thrombin [214,215,216]. Uncontrolled seizures, either idiopathic or related to existing brain injury, lead to increased permeability of the BBB and an associated increase in the concentration of thrombin [61]. These factors trigger a positive feedback loop that promotes epilpetogenesis [217]. Stroke may also precipitate the development of post-stroke epilepsy by generating permanent structural changes from which an epileptic focus arises [112]. Additionally, thrombin has direct epileptogenic effects. Thrombin triggers the generation of epileptic seizures by increasing excitatory tone and decreasing inhibition in CA3 neurons of the hippocampus [87]. This process is mediated by PAR1 [89]. Activation may occur in a manner independent of NMDA by amplifying persistent voltage-gated sodium channel current through tetrodotoxin-sensitive channels [218]. Increased thrombin immunofluorescence is visualized in the hippocampus of mice with pilocarpine-induced status epilepticus, while systemic inhibition of thrombin mitigates the behavioral outcome of pilocarpine in this model [219]. Although investigators have suggested that oral anticoagulants that target thrombin and PAR1 may be useful anti-epileptic medications [219], continued research is necessary.

9. Primary CNS Tumors

The involvement of thrombin in primary CNS tumors has also been increasingly studied. Generally, thrombin promotes tumor cell adhesion, enhances tumor cell growth, upregulates tumor-related angiogenesis, and increases tumor cell seeding and metastases [38,220,221]. This is also the case in primary CNS tumors [222]. Thrombin activity is increased in high-grade glioma and non-glial malignant CNS tumor cell lines [223]. Meizothrombin stimulates human glioblastoma cells via interaction with PAR1 [224]. Tumor precursor cells derived from primary human gliomas and glioblastoma cells overexpress PAR1 [225,226,227]. PAR-1 is involved in thrombin-induced Ca2+ mobilization in human meningioma cells [228]. Similarly, PAR1 inhibition suppresses self-renewal and growth of glioma progenitor cells and gliomas in vivo [225]. PAR1 also mediates protein kinase A (PKA) activation via the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB)-associated catalytic protein kinase A α subunit rather than cAMP levels in glioblastoma cells [229]. Interestingly, other factors including platelet-derived growth factor may be involved in the pathogenesis of glioblastoma [230]. Thrombin-induced A172 human glioblastoma cell proliferation has been found to depend on the level of platelet-derived growth factor-AB (PDGF-AB) [230]. Anti-PAR1 antibodies do not affect the secretion of PDGF-AB or cell growth [230]. Elevated thrombin activity is associated with brain edema resulting from tumor-induced breakdown of the BBB [223]. These pathomechanisms may lead to the formalization of biomarkers for glioblastoma based on components of the thrombin pathway, such as anti-thrombin [231]. A novel six amino acid chloromethyl-ketone compound that inhibits PAR1 activation decreases glioblastoma proliferation rate, colony formation, and invasion in vitro and increased survival and reduced edema volume formation in rats [232]. Similarly, dabigatran inhibits growth, cell cycle progression, migration, and formation of endothelial tubes in glioblastoma cells [233]. Argatroban reduces glioma mass and prolongs survival [234]. Although additional investigation is required, the thrombin pathway represents a potential target for novel therapeutic strategies in humans.

10. Conclusions

Thrombin is involved in coagulation, inflammation, cell protection, and apoptosis. Increasingly, the role of thrombin in the CNS has been explored. Although low concentrations of thrombin are neuroprotective, high concentrations of thrombin exert pathological effects in the CNS through BBB disruption, neuroinflammation, neurotoxicity, maladaptive synaptic plasticity, and coagulation. PAR1 is predominantly responsible for modulating these effects. Involvement of thrombin in neurodegenerative diseases and intracerebral hemorrhage is reasonably well characterized. There is tremendous potential for application of the knowledge regarding thrombin to the management of stroke, particularly with the increased usage of endovascular thrombectomy clot retrieval and the development of novel oral anticoagulants. The roles of thrombin in CNS infections, TBI, epilepsy, and primary CNS tumors continue to be elucidated. Further research will clarify the mechanisms of thrombin pathogenesis in these conditions to catalyze the generation of appropriate therapeutics.

Author Contributions

Conceptualization, N.A.S., M.H.-E. and C.G.P.; methodology, N.A.S.; validation, Z.I.-H., S.H. and C.G.P.; writing—original draft preparation, N.A.S.; writing—review and editing, M.H.-E.; visualization, Z.I.-H., S.H. and C.G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported in part by the Ari and Regine Aprijaskis Fund; and in part by The Adelson Center for the Biology of Addictive Diseases and Chair Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

No disclosures.

References

  1. Di Cera, E. Thrombin. Mol. Asp. Med. 2008, 29, 203–254. [Google Scholar] [CrossRef] [PubMed]
  2. Di Cera, E.; Page, M.J.; Bah, A.; Bush-Pelc, L.A.; Garvey, L.C. Thrombin allostery. Phys. Chem. Chem. Phys. 2007, 9, 1291–1306. [Google Scholar] [CrossRef] [PubMed]
  3. Wells, C.M.; Di Cera, E. Thrombin is a sodium ion activated enzyme. Biochemistry 1992, 31, 11721–11730. [Google Scholar] [CrossRef]
  4. Posma, J.; Posthuma, J.; Spronk, H. Coagulation and non-coagulation effects of thrombin. J. Thromb. Haemost. 2016, 14, 1908–1916. [Google Scholar] [CrossRef] [Green Version]
  5. Ma, L.; Dorling, A. The roles of thrombin and protease-activated receptors in inflammation. Semin. Immunopathol. 2012, 34, 63–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Crawley, J.; Zanardelli, S.; Chion, C.; Lane, D. The central role of thrombin in hemostasis. J. Thromb. Haemost. 2007, 5, 95–101. [Google Scholar] [CrossRef]
  7. Walker, C.; Royston, D. Thrombin generation and its inhibition: A review of the scientific basis and mechanism of action of anticoagulant therapies. Br. J. Anaesth. 2002, 88, 848–863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Krenzlin, H.; Lorenz, V.; Danckwardt, S.; Kempski, O.; Alessandri, B. The importance of thrombin in cerebral injury and disease. Int. J. Mol. Sci. 2016, 17, 84. [Google Scholar] [CrossRef] [Green Version]
  9. Huntington, J. Molecular recognition mechanisms of thrombin. J. Thromb. Haemost. 2005, 3, 1861–1872. [Google Scholar] [CrossRef] [PubMed]
  10. Coughlin, S.R. How the protease thrombin talks to cells. Proc. Natl. Acad. Sci. USA 1999, 96, 11023–11027. [Google Scholar] [CrossRef] [Green Version]
  11. Donovan, F.M.; Cunningham, D.D. Signaling pathways involved in thrombin-induced cell protection. J. Biol. Chem. 1998, 273, 12746–12752. [Google Scholar] [CrossRef] [Green Version]
  12. Davie, E.W.; Fujikawa, K.; Kisiel, W. The coagulation cascade: Initiation, maintenance, and regulation. Biochemistry 1991, 30, 10363–10370. [Google Scholar] [CrossRef]
  13. Weitz, J.; Hudoba, M.; Massel, D.; Maraganore, J.; Hirsh, J. Clot-bound thrombin is protected from inhibition by heparin-antithrombin III but is susceptible to inactivation by antithrombin III-independent inhibitors. J. Clin. Investig. 1990, 86, 385–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Roemisch, J.; Gray, E.; Hoffmann, J.; Wiedermann, C. Antithrombin: A new look at the actions of a serine protease inhibitor. Blood Coagul. Fibrinolysis 2002, 13, 657–670. [Google Scholar] [CrossRef] [PubMed]
  15. Grütter, M.; Priestle, J.; Rahuel, J.; Grossenbacher, H.; Bode, W.; Hofsteenge, J.; Stone, S. Crystal structure of the thrombin-hirudin complex: A novel mode of serine protease inhibition. EMBO J. 1990, 9, 2361–2365. [Google Scholar] [CrossRef]
  16. Stone, S.R.; Hofsteenge, J. Kinetics of the inhibition of thrombin by hirudin. Biochemistry 1986, 25, 4622–4628. [Google Scholar] [CrossRef] [PubMed]
  17. Siller-Matula, J.M.; Schwameis, M.; Blann, A.; Mannhalter, C.; Jilma, B. Thrombin as a multi-functional enzyme. Thromb. Haemost. 2011, 106, 1020–1033. [Google Scholar]
  18. De Luca, C.; Virtuoso, A.; Maggio, N.; Papa, M. Neuro-coagulopathy: Blood coagulation factors in central nervous system diseases. Int. J. Mol. Sci. 2017, 18, 2128. [Google Scholar] [CrossRef]
  19. Mann, K.; Brummel, K.; Butenas, S. What is all that thrombin for? J. Thromb. Haemost. 2003, 1, 1504–1514. [Google Scholar] [CrossRef]
  20. Spiel, A.O.; Bartko, J.; Schwameis, M.; Firbas, C.; Siller-Matula, J.; Schuetz, M.; Weigl, M.; Jilma, B. Increased platelet aggregation and in vivo platelet activation after granulocyte colony-stimulating factor administration. A randomised controlled trial. Thromb. Haemost. 2011, 105, 655. [Google Scholar] [CrossRef]
  21. Wu, C.-C.; Wang, W.-Y.; Wei, C.-K.; Teng, C.-M. Combined blockade of thrombin anion binding exosite-1 and PAR4 produces synergistic antiplatelet effect in human platelets. Thromb. Haemost. 2011, 105, 88–95. [Google Scholar]
  22. Siller-Matula, J.M.; Krumphuber, J.; Jilma, B. Pharmacokinetic, pharmacodynamic and clinical profile of novel antiplatelet drugs targeting vascular diseases. Br. J. Pharmacol. 2010, 159, 502–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lin, J.H.; Garand, M.; Zagorac, B.; Schadinger, S.L.; Scipione, C.; Koschinsky, M.L.; Boffa, M.B. Identification of human thrombin-activatable fibrinolysis inhibitor in vascular and inflammatory cells. Thromb. Haemost. 2011, 105, 999–1009. [Google Scholar] [CrossRef]
  24. Vorjohann, S.; Fish, R.J.; Biron-Andreani, C.; Nagaswami, C.; Weisel, J.W.; Boulot, P.; Reyftmann, L.; De Moerloose, P.; Neerman-Arbez, M. Hypodysfibrinogenaemia due to production of mutant fibrinogen alpha-chains lacking fibrinopeptide A and polymerisation knob ‘A’. Thromb. Haemost. 2010, 104, 990. [Google Scholar] [CrossRef] [PubMed]
  25. Martorell, L.; Martínez-González, J.; Rodriguez, C.; Gentile, M.; Calvayrac, O.; Badimon, L. Thrombin and protease-activated receptors (PARs) in atherothrombosis. Thromb. Haemost. 2008, 99, 305–315. [Google Scholar] [CrossRef] [PubMed]
  26. Rex, S.; Beaulieu, L.M.; Perlman, D.H.; Vitseva, O.; Blair, P.S.; McComb, M.E.; Costello, C.E.; Freedman, J.E. Immune vs. Thrombotic stimulation of platelets differentially regulates signaling pathways, intracellular protein-protein interactions, and α-granule release. Thromb. Haemost. 2009, 102, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Schuepbach, R.A.; Feistritzer, C.; Fernández, J.A.; Griffin, J.H.; Riewald, M. Protection of vascular barrier integrity by activated protein C in murine models depends on protease-activated receptor-1. Thromb. Haemost. 2009, 101, 724. [Google Scholar]
  28. Chen, L.B.; Buchanan, J.M. Mitogenic activity of blood components. I. Thrombin and prothrombin. Proc. Natl. Acad. Sci. USA 1975, 72, 131–135. [Google Scholar] [CrossRef] [Green Version]
  29. Bar-Shavit, R.; Benezra, M.; Eldor, A.; Hy-Am, E.; Fenton, J., 2nd; Wilner, G.D.; Vlodavsky, I. Thrombin immobilized to extracellular matrix is a potent mitogen for vascular smooth muscle cells: Nonenzymatic mode of action. Cell Regul. 1990, 1, 453–463. [Google Scholar] [CrossRef]
  30. Weiss, R.H.; Maduri, M. The mitogenic effect of thrombin in vascular smooth muscle cells is largely due to basic fibroblast growth factor. J. Biol. Chem. 1993, 268, 5724–5727. [Google Scholar] [CrossRef]
  31. Rabiet, M.-J.P.; Plantier, J.-L.; Rival, Y.; Genoux, Y.; Lampugnani, M.-G.; Dejana, E. Thrombin-induced increase in endothelial permeability is associated with changes in cell-to-cell junction organization. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 488–496. [Google Scholar] [CrossRef]
  32. Garcia, J.; Pavalko, F.; Patterson, C. Vascular endothelial cell activation and permeability responses to thrombin. Blood Coagul. Fibrinolysis Int. J. Haemost. Thromb. 1995, 6, 609–626. [Google Scholar] [CrossRef] [PubMed]
  33. Derkach, D.N.; Ihara, E.; Hirano, K.; Nishimura, J.; Takahashi, S.; Kanaide, H. Thrombin causes endothelium-dependent biphasic regulation of vascular tone in the porcine renal interlobar artery. Br. J. Pharmacol. 2000, 131, 1635–1642. [Google Scholar] [CrossRef] [Green Version]
  34. Rickles, F.R.; Patierno, S.; Fernandez, P.M. Tissue factor, thrombin, and cancer. Chest 2003, 124, 58S–68S. [Google Scholar] [CrossRef]
  35. Kumar, P.; Shen, Q.; Pivetti, C.D.; Lee, E.S.; Wu, M.H.; Yuan, S.Y. Molecular mechanisms of endothelial hyperpermeability: Implications in inflammation. Expert Rev. Mol. Med. 2009, 11, e19. [Google Scholar] [CrossRef] [Green Version]
  36. Petäjä, J. Inflammation and coagulation. An overview. Thromb. Res. 2011, 127, S34–S37. [Google Scholar] [CrossRef]
  37. Rukoyatkina, N.; Begonja, A.J.; Geiger, J.; Eigenthaler, M.; Walter, U.; Gambaryan, S. Phosphatidylserine surface expression and integrin αIIbβ3 activity on thrombin/convulxin stimulated platelets/particles of different sizes. Br. J. Haematol. 2009, 144, 591–602. [Google Scholar] [CrossRef] [PubMed]
  38. Tsopanoglou, N.E.; Maragoudakis, M.E. Role of thrombin in angiogenesis and tumor progression. Semin. Thromb. Hemost. 2004, 30, 63–69. [Google Scholar] [PubMed]
  39. Zhang, P.; Ozdemir, T.; Chung, C.-Y.; Robertson, G.P.; Dong, C. Sequential binding of αVβ3 and ICAM-1 determines fibrin-mediated melanoma capture and stable adhesion to CD11b/CD18 on neutrophils. J. Immunol. 2011, 186, 242–254. [Google Scholar] [CrossRef] [Green Version]
  40. Diebold, I.; Djordjevic, T.; Hess, J.; Görlach, A. Rac-1 promotes pulmonary artery smooth muscle cell proliferation by upregulation of plasminogen activator inhibitor-1: Role of NFκB-dependent hypoxia-inducible factor-1α transcription. Thromb. Haemost. 2008, 100, 1021–1028. [Google Scholar] [CrossRef] [PubMed]
  41. Diebold, I.; Petry, A.; Djordjevic, T.; BelAiba, R.S.; Fineman, J.; Black, S.; Schreiber, C.; Fratz, S.; Hess, J.; Kietzmann, T. Reciprocal regulation of Rac1 and PAK-1 by HIF-1α: A positive-feedback loop promoting pulmonary vascular remodeling. Antioxid. Redox Signal. 2010, 13, 399–412. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, D.; Dorling, A. Critical roles for thrombin in acute and chronic inflammation. J. Thromb. Haemost. 2009, 7, 122–126. [Google Scholar] [CrossRef] [PubMed]
  43. Miyamoto, M.; Ohno, M.; Yamada, N.; Ohtake, A.; Matsushita, T. TRA-418, a thromboxane A2 receptor antagonist and prostacyclin receptor agonist, inhibits platelet-leukocyte interaction in human whole blood. Thromb. Haemost. 2010, 104, 788–795. [Google Scholar] [PubMed]
  44. Verkleij, C.J.; Roelofs, J.J.; Havik, S.R.; Meijers, J.C.; Marx, P.F. The role of thrombin-activatable fibrinolysis inhibitor in diabetic wound healing. Thromb. Res. 2010, 126, 442–446. [Google Scholar] [CrossRef] [PubMed]
  45. Naldini, A.; Carney, D.H.; Pucci, A.; Pasquali, A.; Carraro, F. Thrombin regulates the expression of proangiogenic cytokines via proteolytic activation of protease-activated receptor-1. Gen. Pharmacol. Vasc. Syst. 2000, 35, 255–259. [Google Scholar] [CrossRef]
  46. Bae, J.S.; Kim, Y.U.; Park, M.K.; Rezaie, A.R. Concentration dependent dual effect of thrombin in endothelial cells via Par-1 and Pi3 Kinase. J. Cell. Physiol. 2009, 219, 744–751. [Google Scholar] [CrossRef] [Green Version]
  47. Donovan, F.M.; Pike, C.J.; Cotman, C.W.; Cunningham, D.D. Thrombin induces apoptosis in cultured neurons and astrocytes via a pathway requiring tyrosine kinase and RhoA activities. J. Neurosci. 1997, 17, 5316–5326. [Google Scholar] [CrossRef]
  48. Zampatis, D.E.; Rutz, C.; Furkert, J.; Schmidt, A.; Wüstenhagen, D.; Kubick, S.; Tsopanoglou, N.E.; Schülein, R. The protease-activated receptor 1 possesses a functional and cleavable signal peptide which is necessary for receptor expression. FEBS Lett. 2012, 586, 2351–2359. [Google Scholar] [CrossRef]
  49. O’Brien, P.J.; Molino, M.; Kahn, M.; Brass, L.F. Protease activated receptors: Theme and variations. Oncogene 2001, 20, 1570–1581. [Google Scholar] [CrossRef] [Green Version]
  50. Abraham, L.A.; Mackie, E.J. Modulation of osteoblast-like cell behavior by activation of protease-activated receptor-1. J. Bone Miner. Res. 1999, 14, 1320–1329. [Google Scholar] [CrossRef]
  51. Lin, H.; Liu, A.P.; Smith, T.H.; Trejo, J. Cofactoring and dimerization of proteinase-activated receptors. Pharmacol. Rev. 2013, 65, 1198–1213. [Google Scholar] [CrossRef] [Green Version]
  52. Bae, J.-S.; Yang, L.; Manithody, C.; Rezaie, A.R. The ligand occupancy of endothelial protein C receptor switches the protease-activated receptor 1-dependent signaling specificity of thrombin from a permeability-enhancing to a barrier-protective response in endothelial cells. Blood J. Am. Soc. Hematol. 2007, 110, 3909–3916. [Google Scholar] [CrossRef]
  53. Bea, F.; Kreuzer, J.; Preusch, M.; Schaab, S.; Isermann, B.; Rosenfeld, M.E.; Katus, H.; Blessing, E. Melagatran reduces advanced atherosclerotic lesion size and may promote plaque stability in Apolipoprotein E–deficient mice. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2787–2792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Griffin, J.H.; Zlokovic, B.V.; Mosnier, L.O. Activated protein C: Biased for translation. Blood 2015, 125, 2898–2907. [Google Scholar] [CrossRef] [Green Version]
  55. Soh, U.J.; Dores, M.R.; Chen, B.; Trejo, J. Signal transduction by protease-activated receptors. Br. J. Pharmacol. 2010, 160, 191–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Rezaie, A.R. The occupancy of endothelial protein C receptor by its ligand modulates the par-1 dependent signaling specificity of coagulation proteases. IUBMB Life 2011, 63, 390–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Tohgo, A.; Pierce, K.L.; Choy, E.W.; Lefkowitz, R.J.; Luttrell, L.M. β-Arrestin scaffolding of the ERK cascade enhances cytosolic ERK activity but inhibits ERK-mediated transcription following angiotensin AT1a receptor stimulation. J. Biol. Chem. 2002, 277, 9429–9436. [Google Scholar] [CrossRef] [Green Version]
  58. Tohgo, A.; Choy, E.W.; Gesty-Palmer, D.; Pierce, K.L.; Laporte, S.; Oakley, R.H.; Caron, M.G.; Lefkowitz, R.J.; Luttrell, L.M. The stability of the G protein-coupled receptor-β-arrestin interaction determines the mechanism and functional consequence of ERK activation. J. Biol. Chem. 2003, 278, 6258–6267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Shenoy, S.K.; Barak, L.S.; Xiao, K.; Ahn, S.; Berthouze, M.; Shukla, A.K.; Luttrell, L.M.; Lefkowitz, R.J. Ubiquitination of β-arrestin links seven-transmembrane receptor endocytosis and ERK activation. J. Biol. Chem. 2007, 282, 29549–29562. [Google Scholar] [CrossRef] [Green Version]
  60. Steen, A.; Larsen, O.; Thiele, S.; Rosenkilde, M.M. Biased and g protein-independent signaling of chemokine receptors. Front. Immunol. 2014, 5, 277. [Google Scholar] [CrossRef] [Green Version]
  61. Sokolova, E.; Reiser, G. Prothrombin/thrombin and the thrombin receptors PAR-1 and PAR-4 in the brain: Localization, expression and participation in neurodegenerative diseases. Thromb. Haemost. 2008, 100, 576–581. [Google Scholar] [CrossRef]
  62. Choi, B.; Suzuki, M.; Kim, T.; Wagner, S.; Cunningham, D. Protease nexin-1. Localization in the human brain suggests a protective role against extravasated serine proteases. Am. J. Pathol. 1990, 137, 741. [Google Scholar]
  63. Deschepper, C.F.; Bigornia, V.; Berens, M.E.; Lapointe, M.C. Production of thrombin and antithrombin III by brain and astroglial cell cultures. Mol. Brain Res. 1991, 11, 355–358. [Google Scholar] [CrossRef]
  64. Niclou, S.P.; Suidan, H.S.; Pavlik, A.; Vejsada, R.; Monard, D. Changes in the expression of protease-activated receptor 1 and protease nexin-1 mRNA during rat nervous system development and after nerve lesion. Eur. J. Neurosci. 1998, 10, 1590–1607. [Google Scholar] [CrossRef] [PubMed]
  65. Shikamoto, Y.; Morita, T. Expression of factor X in both the rat brain and cells of the central nervous system. FEBS Lett. 1999, 463, 387–389. [Google Scholar] [CrossRef] [Green Version]
  66. Dihanich, M.; Kaser, M.; Reinhard, E.; Cunningham, D.; Monard, D. Prothrombin mRNA is expressed by cells of the nervous system. Neuron 1991, 6, 575–581. [Google Scholar] [CrossRef]
  67. Ben Shimon, M.; Lenz, M.; Ikenberg, B.; Becker, D.; Shavit Stein, E.; Chapman, J.; Tanne, D.; Pick, C.G.; Blatt, I.; Neufeld, M. Thrombin regulation of synaptic transmission and plasticity: Implications for health and disease. Front. Cell. Neurosci. 2015, 9, 151. [Google Scholar] [CrossRef] [Green Version]
  68. Xi, G.; Reiser, G.; Keep, R.F. The role of thrombin and thrombin receptors in ischemic, hemorrhagic and traumatic brain injury: Deleterious or protective? J. Neurochem. 2003, 84, 3–9. [Google Scholar] [CrossRef] [PubMed]
  69. Stein, E.S.; Itsekson-Hayosh, Z.; Aronovich, A.; Reisner, Y.; Bushi, D.; Pick, C.G.; Tanne, D.; Chapman, J.; Vlachos, A.; Maggio, N. Thrombin induces ischemic LTP (iLTP): Implications for synaptic plasticity in the acute phase of ischemic stroke. Sci. Rep. 2015, 5, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Ubl, J.; Vöhringer, C.; Reiser, G. Co-existence of two types of [Ca2+] i-inducing protease-activated receptors (PAR-1 and PAR-2) in rat astrocytes and C6 glioma cells. Neuroscience 1998, 86, 597–609. [Google Scholar] [CrossRef]
  71. Striggow, F.; Riek-Burchardt, M.; Kiesel, A.; Schmidt, W.; Henrich-Noack, P.; Breder, J.; Krug, M.; Reymann, K.G.; Reiser, G. Four different types of protease-activated receptors are widely expressed in the brain and are up-regulated in hippocampus by severe ischemia. Eur. J. Neurosci. 2001, 14, 595–608. [Google Scholar] [CrossRef]
  72. Junge, C.E.; Lee, C.J.; Hubbard, K.B.; Zhang, Z.; Olson, J.J.; Hepler, J.R.; Brat, D.J.; Traynelis, S.F. Protease-activated receptor-1 in human brain: Localization and functional expression in astrocytes. Exp. Neurol. 2004, 188, 94–103. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, H.; Ubl, J.J.; Reiser, G. Four subtypes of protease-activated receptors, co-expressed in rat astrocytes, evoke different physiological signaling. Glia 2002, 37, 53–63. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, H.; Ubl, J.J.; Stricker, R.; Reiser, G. Thrombin (PAR-1)-induced proliferation in astrocytes via MAPK involves multiple signaling pathways. Am. J. Physiol. Cell Physiol. 2002, 283, C1351–C1364. [Google Scholar] [CrossRef]
  75. Jiang, Y.; Wu, J.; Hua, Y.; Keep, R.F.; Xiang, J.; Hoff, J.T.; Xi, G. Thrombin-receptor activation and thrombin-induced brain tolerance. J. Cereb. Blood Flow Metab. 2002, 22, 404–410. [Google Scholar] [CrossRef]
  76. Jamison, C.S.; Degen, S.J.F. Prenatal and postnatal expression of mRNA coding for rat prothrombin. Biochim. Biophys. Acta Gene Struct. Expr. 1991, 1088, 208–216. [Google Scholar] [CrossRef]
  77. Weinstein, J.; Gold, S.J.; Cunningham, D.D.; Gall, C. Cellular localization of thrombin receptor mRNA in rat brain: Expression by mesencephalic dopaminergic neurons and codistribution with prothrombin mRNA. J. Neurosci. 1995, 15, 2906–2919. [Google Scholar] [CrossRef] [Green Version]
  78. Debeir, T.; Gueugnon, J.; Vigé, X.; Benavides, J. Transduction mechanisms involved in thrombin receptor-induced nerve growth factor secretion and cell division in primary cultures of astrocytes. J. Neurochem. 1996, 66, 2320–2328. [Google Scholar] [CrossRef]
  79. Suo, Z.; Wu, M.; Ameenuddin, S.; Anderson, H.E.; Zoloty, J.E.; Citron, B.A.; Andrade-Gordon, P.; Festoff, B.W. Participation of protease-activated receptor-1 in thrombin-induced microglial activation. J. Neurochem. 2002, 80, 655–666. [Google Scholar] [CrossRef] [PubMed]
  80. Laskowski, A.; Reiser, G.; Reymann, K.G. Protease-activated receptor-1 induces generation of new microglia in the dentate gyrus of traumatised hippocampal slice cultures. Neurosci. Lett. 2007, 415, 17–21. [Google Scholar] [CrossRef]
  81. Pai, K.S.; Mahajan, V.B.; Lau, A.; Cunningham, D.D. Thrombin receptor signaling to cytoskeleton requires Hsp90. J. Biol. Chem. 2001, 276, 32642–32647. [Google Scholar] [CrossRef] [Green Version]
  82. Cunningham, D.D.; Gurwitz, D. Proteolytic regulation of neurite outgrowth from neuroblastoma cells by thrombin and protease nexin-1. J. Cell. Biochem. 1989, 39, 55–64. [Google Scholar] [CrossRef]
  83. Grabham, P.; Cunningham, D.D. Thrombin receptor activation stimulates astrocyte proliferation and reversal of stellation by distinct pathways: Involvement of tyrosine phosphorylation. J. Neurochem. 1995, 64, 583–591. [Google Scholar] [CrossRef]
  84. Wang, H.; Reiser, G. Thrombin signaling in the brain: The role of protease-activated receptors. Biol. Chem. 2003, 384, 193–202. [Google Scholar] [CrossRef]
  85. Jalink, K.; Van Corven, E.J.; Hengeveld, T.; Morii, N.; Narumiya, S.; Moolenaar, W.H. Inhibition of lysophosphatidate-and thrombin-induced neurite retraction and neuronal cell rounding by ADP ribosylation of the small GTP-binding protein Rho. J. Cell Biol. 1994, 126, 801–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jalink, K.; Moolenaar, W.H. Thrombin receptor activation causes rapid neural cell rounding and neurite retraction independent of classic second messengers. J. Cell Biol. 1992, 118, 411–419. [Google Scholar] [CrossRef] [Green Version]
  87. Maggio, N.; Cavaliere, C.; Papa, M.; Blatt, I.; Chapman, J.; Segal, M. Thrombin regulation of synaptic transmission: Implications for seizure onset. Neurobiol. Dis. 2013, 50, 171–178. [Google Scholar] [CrossRef] [PubMed]
  88. Maggio, N.; Itsekson, Z.; Dominissini, D.; Blatt, I.; Amariglio, N.; Rechavi, G.; Tanne, D.; Chapman, J. Thrombin regulation of synaptic plasticity: Implications for physiology and pathology. Exp. Neurol. 2013, 247, 595–604. [Google Scholar] [CrossRef]
  89. Maggio, N.; Shavit, E.; Chapman, J.; Segal, M. Thrombin induces long-term potentiation of reactivity to afferent stimulation and facilitates epileptic seizures in rat hippocampal slices: Toward understanding the functional consequences of cerebrovascular insults. J. Neurosci. 2008, 28, 732–736. [Google Scholar] [CrossRef] [PubMed]
  90. Lee, C.J.; Mannaioni, G.; Yuan, H.; Woo, D.H.; Gingrich, M.B.; Traynelis, S.F. Astrocytic control of synaptic NMDA receptors. J. Physiol. 2007, 581, 1057–1081. [Google Scholar] [CrossRef] [PubMed]
  91. Gingrich, M.B.; Junge, C.E.; Lyuboslavsky, P.; Traynelis, S.F. Potentiation of NMDA receptor function by the serine protease thrombin. J. Neurosci. 2000, 20, 4582–4595. [Google Scholar] [CrossRef] [PubMed]
  92. Almonte, A.G.; Hamill, C.E.; Chhatwal, J.P.; Wingo, T.S.; Barber, J.A.; Lyuboslavsky, P.N.; Sweatt, J.D.; Ressler, K.J.; White, D.A.; Traynelis, S.F. Learning and memory deficits in mice lacking protease activated receptor-1. Neurobiol. Learn. Mem. 2007, 88, 295–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hamill, C.E.; Mannaioni, G.; Lyuboslavsky, P.; Sastre, A.A.; Traynelis, S.F. Protease-activated receptor 1-dependent neuronal damage involves NMDA receptor function. Exp. Neurol. 2009, 217, 136–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mannaioni, G.; Orr, A.G.; Hamill, C.E.; Yuan, H.; Pedone, K.H.; McCoy, K.L.; Palmini, R.B.; Junge, C.E.; Lee, C.J.; Yepes, M. Plasmin potentiates synaptic N-methyl-D-aspartate receptor function in hippocampal neurons through activation of protease-activated receptor-1. J. Biol. Chem. 2008, 283, 20600–20611. [Google Scholar] [CrossRef] [Green Version]
  95. Han, K.-S.; Mannaioni, G.; Hamill, C.E.; Lee, J.; Junge, C.E.; Lee, C.J.; Traynelis, S.F. Activation of protease activated receptor 1 increases the excitability of the dentate granule neurons of hippocampus. Mol. Brain 2011, 4, 1–12. [Google Scholar] [CrossRef] [Green Version]
  96. Sinnreich, M.; Meins, M.; Niclou, S.P.; Suidan, H.S.; Monard, D. Prothrombin overexpressed in post-natal neurones requires blood factors for activation in the mouse brain. J. Neurochem. 2004, 88, 1380–1388. [Google Scholar] [CrossRef]
  97. Brailoiu, E.; Shipsky, M.M.; Yan, G.; Abood, M.E.; Brailoiu, G.C. Mechanisms of modulation of brain microvascular endothelial cells function by thrombin. Brain Res. 2017, 1657, 167–175. [Google Scholar] [CrossRef] [Green Version]
  98. Machida, T.; Takata, F.; Matsumoto, J.; Takenoshita, H.; Kimura, I.; Yamauchi, A.; Dohgu, S.; Kataoka, Y. Brain pericytes are the most thrombin-sensitive matrix metalloproteinase-9-releasing cell type constituting the blood–brain barrier in vitro. Neurosci. Lett. 2015, 599, 109–114. [Google Scholar] [CrossRef]
  99. Li, L.; Tao, Y.; Tang, J.; Chen, Q.; Yang, Y.; Feng, Z.; Chen, Y.; Yang, L.; Yang, Y.; Zhu, G. A cannabinoid receptor 2 agonist prevents thrombin-induced blood–brain barrier damage via the inhibition of microglial activation and matrix metalloproteinase expression in rats. Transl. Stroke Res. 2015, 6, 467–477. [Google Scholar] [CrossRef] [PubMed]
  100. Machida, T.; Dohgu, S.; Takata, F.; Matsumoto, J.; Kimura, I.; Koga, M.; Nakamoto, K.; Yamauchi, A.; Kataoka, Y. Role of thrombin-PAR1-PKCθ/δ axis in brain pericytes in thrombin-induced MMP-9 production and blood–brain barrier dysfunction in vitro. Neuroscience 2017, 350, 146–157. [Google Scholar] [CrossRef]
  101. Nishino, A.; Suzuki, M.; Ohtani, H.; Motohashi, O.; Umezawa, K.; Nagura, H.; Yoshimoto, T. Thrombin may contribute to the pathophysiology of central nervous system injury. J. Neurotrauma 1993, 10, 167–179. [Google Scholar] [CrossRef]
  102. Wang, Y.; Luo, W.; Reiser, G. Activation of protease-activated receptors in astrocytes evokes a novel neuroprotective pathway through release of chemokines of the growth-regulated oncogene/cytokine-induced neutrophil chemoattractant family. Eur. J. Neurosci. 2007, 26, 3159–3168. [Google Scholar] [CrossRef]
  103. Ryu, J.; Pyo, H.; Jou, I.; Joe, E. Thrombin induces NO release from cultured rat microglia via protein kinase C, mitogen-activated protein kinase, and NF-κB. J. Biol. Chem. 2000, 275, 29955–29959. [Google Scholar] [CrossRef] [Green Version]
  104. Li, D.-Q.; Zhou, Y.-P.; Yang, H. Donepezil combined with natural hirudin improves the clinical symptoms of patients with mild-to-moderate Alzheimer’s disease: A 20-week open-label pilot study. Int. J. Med Sci. 2012, 9, 248. [Google Scholar] [CrossRef] [Green Version]
  105. Fujimoto, S.; Katsuki, H.; Kume, T.; Akaike, A. Thrombin-induced delayed injury involves multiple and distinct signaling pathways in the cerebral cortex and the striatum in organotypic slice cultures. Neurobiol. Dis. 2006, 22, 130–142. [Google Scholar] [CrossRef] [Green Version]
  106. Fujimoto, S.; Katsuki, H.; Ohnishi, M.; Takagi, M.; Kume, T.; Akaike, A. Thrombin induces striatal neurotoxicity depending on mitogen-activated protein kinase pathways in vivo. Neuroscience 2007, 144, 694–701. [Google Scholar] [CrossRef] [Green Version]
  107. Xue, M.; Hollenberg, M.D.; Yong, V.W. Combination of thrombin and matrix metalloproteinase-9 exacerbates neurotoxicity in cell culture and intracerebral hemorrhage in mice. J. Neurosci. 2006, 26, 10281–10291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Xue, M.; Fan, Y.; Liu, S.; Zygun, D.A.; Demchuk, A.; Yong, V.W. Contributions of multiple proteases to neurotoxicity in a mouse model of intracerebral haemorrhage. Brain 2009, 132, 26–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Marques, M.A.; Tolar, M.; Harmony, J.; Crutcher, K.A. A thrombin cleavage fragment of apolipoprotein E exhibits isoform-specific neurotoxicity. Neuroreport 1996, 7, 2529–2532. [Google Scholar] [CrossRef] [PubMed]
  110. Tolar, M.; Marques, M.A.; Harmony, J.A.; Crutcher, K.A. Neurotoxicity of the 22 kDa thrombin-cleavage fragment of apolipoprotein E and related synthetic peptides is receptor-mediated. J. Neurosci. 1997, 17, 5678–5686. [Google Scholar] [CrossRef]
  111. Grammas, P.; Ottman, T.; Reimann-Philipp, U.; Larabee, J.; Weigel, P.H. Injured brain endothelial cells release neurotoxic thrombin. J. Alzheimer’s Dis. 2004, 6, 275–281. [Google Scholar] [CrossRef] [PubMed]
  112. Altman, K.; Shavit Stein, E.; Maggio, N. Post stroke seizures and epilepsy: From proteases to maladaptive plasticity. Front. Cell. Neurosci. 2019, 13, 397. [Google Scholar] [CrossRef] [Green Version]
  113. Lee, K.R.; Colon, G.P.; Betz, A.L.; Keep, R.F.; Kim, S.; Hoff, J.T. Edema from intracerebral hemorrhage: The role of thrombin. J. Neurosurg. 1996, 84, 91–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Akiyama, H.; Ikeda, K.; Kondo, H.; McGeer, P.L. Thrombin accumulation in brains of patients with Alzheimer’s disease. Neurosci. Lett. 1992, 146, 152–154. [Google Scholar] [CrossRef]
  115. Arai, T.; Miklossy, J.; Klegeris, A.; Guo, J.-P.; McGeer, P.L. Thrombin and prothrombin are expressed by neurons and glial cells and accumulate in neurofibrillary tangles in Alzheimer disease brain. J. Neuropathol. Exp. Neurol. 2006, 65, 19–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Pompili, E.; Nori, S.L.; Geloso, M.C.; Guadagni, E.; Corvino, V.; Michetti, F.; Fumagalli, L. Trimethyltin-induced differential expression of PAR subtypes in reactive astrocytes of the rat hippocampus. Mol. Brain Res. 2004, 122, 93–98. [Google Scholar] [CrossRef]
  117. Zamolodchikov, D.; Renné, T.; Strickland, S. The Alzheimer’s disease peptide β-amyloid promotes thrombin generation through activation of coagulation factor XII. J. Thromb. Haemost. 2016, 14, 995–1007. [Google Scholar] [CrossRef] [Green Version]
  118. Grammas, P.; Samany, P.G.; Thirumangalakudi, L. Thrombin and inflammatory proteins are elevated in Alzheimer’s disease microvessels: Implications for disease pathogenesis. J. Alzheimer’s Dis. 2006, 9, 51–58. [Google Scholar] [CrossRef]
  119. van der Poll, T.; Büller, H.R.; ten Cate, H.; Wortel, C.H.; Bauer, K.A.; van Deventer, S.J.; Hack, C.E.; Sauerwein, H.P.; Rosenberg, R.D.; ten Cate, J.W. Activation of coagulation after administration of tumor necrosis factor to normal subjects. New Engl. J. Med. 1990, 322, 1622–1627. [Google Scholar] [CrossRef] [PubMed]
  120. Iannucci, J.; Renehan, W.; Grammas, P. Thrombin, a Mediator of Coagulation, Inflammation, and Neurotoxicity at the Neurovascular Interface: Implications for Alzheimer’s Disease. Front. Neurosci. 2020, 14, 762. [Google Scholar] [CrossRef]
  121. Suo, Z.; Wu, M.; Citron, B.A.; Palazzo, R.E.; Festoff, B.W. Rapid tau aggregation and delayed hippocampal neuronal death induced by persistent thrombin signaling. J. Biol. Chem. 2003, 278, 37681–37689. [Google Scholar] [CrossRef] [Green Version]
  122. Suo, Z.; Wu, M.; Citron, B.A.; Gao, C.; Festoff, B.W. Persistent protease-activated receptor 4 signaling mediates thrombin-induced microglial activation. J. Biol. Chem. 2003, 278, 31177–31183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Davis-Salinas, J.; Saporito-Irwin, S.M.; Donovan, F.M.; Cunningham, D.D.; Van Nostrand, W. Thrombin receptor activation induces secretion and nonamyloidogenic processing of amyloid beta-protein precursor. J. Biol. Chem. 1994, 269, 22623–22627. [Google Scholar] [CrossRef]
  124. Brewer, G.J. Thrombin Causes Cell Spreading and Redistribution of β-Amyloid Immunoreactivity in Cultured Hippocampal Neurons. J. Neurochem. 1996, 67, 119–130. [Google Scholar] [CrossRef] [PubMed]
  125. Smith-Swintosky, V.L.; Zimmer, S.; Fenton, J.W.; Mattson, M.P. Opposing Actions of Thrombin and Protease Nexin-1 on Amyloid β-Peptide Toxicity and on Accumulation of Peroxides and Calcium in Hippocampal Neurons. J. Neurochem. 1995, 65, 1415–1418. [Google Scholar] [CrossRef]
  126. Pike, C.J.; Vaughan, P.J.; Cunningham, D.D.; Cotman, C.W. Thrombin attenuates neuronal cell death and modulates astrocyte reactivity induced by β-amyloid in vitro. J. Neurochem. 1996, 66, 1374–1382. [Google Scholar] [CrossRef]
  127. Vaughan, P.J.; Su, J.; Cotman, C.W.; Cunningham, D.D. Protease nexin-1, a potent thrombin inhibitor, is reduced around cerebral blood vessels in Alzheimer’s disease. Brain Res. 1994, 668, 160–170. [Google Scholar] [CrossRef]
  128. Choi, S.-H.; Lee, D.Y.; Kim, S.U.; Jin, B.K. Thrombin-induced oxidative stress contributes to the death of hippocampal neurons in vivo: Role of microglial NADPH oxidase. J. Neurosci. 2005, 25, 4082–4090. [Google Scholar] [CrossRef]
  129. Iannucci, J.; Johnson, S.L.; Majchrzak, M.; Barlock, B.J.; Akhlaghi, F.; Seeram, N.P.; Sen, A.; Grammas, P. Short-term treatment with dabigatran alters protein expression patterns in a late-stage tau-based Alzheimer’s disease mouse model. Biochem. Biophys. Rep. 2020, 24, 100862. [Google Scholar]
  130. Tripathy, D.; Sanchez, A.; Yin, X.; Luo, J.; Martinez, J.; Grammas, P. Thrombin, a mediator of cerebrovascular inflammation in AD and hypoxia. Front. Aging Neurosci. 2013, 5, 19. [Google Scholar] [CrossRef] [Green Version]
  131. Rami, B.K. Direct thrombin inhibitors’ potential efficacy in Alzheimer’s disease. Am. J. Alzheimer’s Dis. Other Dement. 2012, 27, 564–567. [Google Scholar] [CrossRef] [PubMed]
  132. Choi, S.-H.; Lee, D.Y.; Ryu, J.K.; Kim, J.; Joe, E.H.; Jin, B.K. Thrombin induces nigral dopaminergic neurodegeneration in vivo by altering expression of death-related proteins. Neurobiol. Dis. 2003, 14, 181–193. [Google Scholar] [CrossRef]
  133. Carreño-Müller, E.; Herrera, A.J.; De Pablos, R.M.; Tomás-Camardiel, M.; Venero, J.L.; Cano, J.; Machado, A. Thrombin induces in vivo degeneration of nigral dopaminergic neurones along with the activation of microglia. J. Neurochem. 2003, 84, 1201–1214. [Google Scholar] [CrossRef]
  134. Lee, D.Y.; Oh, Y.J.; Jin, B.K. Thrombin-activated microglia contribute to death of dopaminergic neurons in rat mesencephalic cultures: Dual roles of mitogen-activated protein kinase signaling pathways. Glia 2005, 51, 98–110. [Google Scholar] [CrossRef] [PubMed]
  135. Katsuki, H.; Okawara, M.; Shibata, H.; Kume, T.; Akaike, A. Nitric oxide-producing microglia mediate thrombin-induced degeneration of dopaminergic neurons in rat midbrain slice culture. J. Neurochem. 2006, 97, 1232–1242. [Google Scholar] [CrossRef] [Green Version]
  136. Choi, S.-H.; Joe, E.H.; Kim, S.U.; Jin, B.K. Thrombin-induced microglial activation produces degeneration of nigral dopaminergic neurons in vivo. J. Neurosci. 2003, 23, 5877–5886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Cannon, J.R.; Hua, Y.; Richardson, R.J.; Xi, G.; Keep, R.F.; Schallert, T. The effect of thrombin on a 6-hydroxydopamine model of Parkinson’s disease depends on timing. Behav. Brain Res. 2007, 183, 161–168. [Google Scholar] [CrossRef] [Green Version]
  138. Jenner, P. Oxidative stress in Parkinson’s disease. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2003, 53, S26–S38. [Google Scholar] [CrossRef] [PubMed]
  139. Ishida, Y.; Nagai, A.; Kobayashi, S.; Kim, S.U. Upregulation of protease-activated receptor-1 in astrocytes in Parkinson disease: Astrocyte-mediated neuroprotection through increased levels of glutathione peroxidase. J. Neuropathol. Exp. Neurol. 2006, 65, 66–77. [Google Scholar] [CrossRef] [Green Version]
  140. Cannon, J.R.; Keep, R.F.; Schallert, T.; Hua, Y.; Richardson, R.J.; Xi, G. Protease-activated receptor-1 mediates protection elicited by thrombin preconditioning in a rat 6-hydroxydopamine model of Parkinson’s disease. Brain Res. 2006, 1116, 177–186. [Google Scholar] [CrossRef]
  141. Cannon, J.R.; Keep, R.F.; Hua, Y.; Richardson, R.J.; Schallert, T.; Xi, G. Thrombin preconditioning provides protection in a 6-hydroxydopamine Parkinson’s disease model. Neurosci. Lett. 2005, 373, 189–194. [Google Scholar] [CrossRef]
  142. Kandil, E.A.; Sayed, R.H.; Ahmed, L.A.; Abd El Fattah, M.A.; El-Sayeh, B.M. Modulatory role of Nurr1 activation and thrombin inhibition in the neuroprotective effects of dabigatran etexilate in rotenone-induced Parkinson’s disease in rats. Mol. Neurobiol. 2018, 55, 4078–4089. [Google Scholar] [CrossRef]
  143. Johnson, S.L.; Iannucci, J.; Seeram, N.P.; Grammas, P. Inhibiting thrombin improves motor function and decreases oxidative stress in the LRRK2 transgenic Drosophila melanogaster model of Parkinson’s disease. Biochem. Biophys. Res. Commun. 2020, 527, 532–538. [Google Scholar] [CrossRef]
  144. Pretorius, E.; Page, M.J.; Mbotwe, S.; Kell, D.B. Lipopolysaccharide-binding protein (LBP) can reverse the amyloid state of fibrin seen or induced in Parkinson’s disease. PLoS ONE 2018, 13, e0192121. [Google Scholar] [CrossRef] [Green Version]
  145. Pretorius, E.; Mbotwe, S.; Bester, J.; Robinson, C.J.; Kell, D.B. Acute induction of anomalous and amyloidogenic blood clotting by molecular amplification of highly substoichiometric levels of bacterial lipopolysaccharide. J. R. Soc. Interface 2016, 13, 20160539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Paterson, P.; Koh, C.; Kwaan, H. Role of the clotting system in the pathogenesis of neuroimmunologic disease. Fed. Proc. 1987, 46, 91. [Google Scholar]
  147. Davalos, D.; Baeten, K.M.; Whitney, M.A.; Mullins, E.S.; Friedman, B.; Olson, E.S.; Ryu, J.K.; Smirnoff, D.S.; Petersen, M.A.; Bedard, C. Early detection of thrombin activity in neuroinflammatory disease. Ann. Neurol. 2014, 75, 303–308. [Google Scholar] [CrossRef] [Green Version]
  148. Constantinescu, C.S.; Farooqi, N.; O’Brien, K.; Gran, B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br. J. Pharmacol. 2011, 164, 1079–1106. [Google Scholar] [CrossRef] [PubMed]
  149. Beilin, O.; Karussis, D.M.; Korczyn, A.D.; Gurwitz, D.; Aronovich, R.; Hantai, D.; Grigoriadis, N.; Mizrachi-Kol, R.; Chapman, J. Increased thrombin inhibition in experimental autoimmune encephalomyelitis. J. Neurosci. Res. 2005, 79, 351–359. [Google Scholar] [CrossRef] [PubMed]
  150. Inaba, Y.; Ichikawa, M.; Inoue, A.; Itoh, M.; Kyogashima, M.; Sekiguchi, Y.; Nakamura, S.; Komiyama, A.; Koh, C.-S. Plasma thrombin–antithrombin III complex is associated with the severity of experimental autoimmune encephalomyelitis. J. Neurol. Sci. 2001, 185, 89–93. [Google Scholar] [CrossRef]
  151. Göbel, K.; Pankratz, S.; Asaridou, C.-M.; Herrmann, A.M.; Bittner, S.; Merker, M.; Ruck, T.; Glumm, S.; Langhauser, F.; Kraft, P. Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells. Nat. Commun. 2016, 7, 11626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Göbel, K.; Eichler, S.; Wiendl, H.; Chavakis, T.; Kleinschnitz, C.; Meuth, S.G. The coagulation factors fibrinogen, thrombin, and factor XII in inflammatory disorders—A systematic review. Front. Immunol. 2018, 9, 1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Dziedzic, A.; Miller, E.; Bijak, M.; Przyslo, L.; Saluk-Bijak, J. Increased Pro-Thrombotic Platelet Activity Associated with Thrombin/PAR1-Dependent Pathway Disorder in Patients with Secondary Progressive Multiple Sclerosis. Int. J. Mol. Sci. 2020, 21, 7722. [Google Scholar] [CrossRef]
  154. Wang, Y.; Richter-Landsberg, C.; Reiser, G. Expression of protease-activated receptors (PARs) in OLN-93 oligodendroglial cells and mechanism of PAR-1-induced calcium signaling. Neuroscience 2004, 126, 69–82. [Google Scholar] [CrossRef]
  155. Noorbakhsh, F.; Tsutsui, S.; Vergnolle, N.; Boven, L.A.; Shariat, N.; Vodjgani, M.; Warren, K.G.; Andrade-Gordon, P.; Hollenberg, M.D.; Power, C. Proteinase-activated receptor 2 modulates neuroinflammation in experimental autoimmune encephalomyelitis and multiple sclerosis. J. Exp. Med. 2006, 203, 425–435. [Google Scholar] [CrossRef]
  156. Juhler, M.; Barry, D.I.; Offner, H.; Konat, G.; Klinken, L.; Paulson, O.B. Blood-brain and blood-spinal cord barrier permeability during the course of experimental allergic encephalomyelitis in the rat. Brain Res. 1984, 302, 347–355. [Google Scholar] [CrossRef]
  157. Kim, H.N.; Kim, Y.R.; Ahn, S.M.; Lee, S.K.; Shin, H.K.; Choi, B.T. Protease activated receptor-1 antagonist ameliorates the clinical symptoms of experimental autoimmune encephalomyelitis via inhibiting breakdown of blood–brain barrier. J. Neurochem. 2015, 135, 577–588. [Google Scholar] [CrossRef]
  158. Verbout, N.G.; Yu, X.; Healy, L.D.; Phillips, K.G.; Tucker, E.I.; Gruber, A.; McCarty, O.J.; Offner, H. Thrombin mutant W215A/E217A treatment improves neurological outcome and attenuates central nervous system damage in experimental autoimmune encephalomyelitis. Metab. Brain Dis. 2015, 30, 57–65. [Google Scholar] [CrossRef] [Green Version]
  159. Carcaillon, L.; Alhenc-Gelas, M.; Bejot, Y.; Spaft, C.; Ducimetière, P.; Ritchie, K.; Dartigues, J.-F.; Scarabin, P.-Y. Increased thrombin generation is associated with acute ischemic stroke but not with coronary heart disease in the elderly: The Three-City cohort study. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1445–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Riek-Burchardt, M.; Striggow, F.; Henrich-Noack, P.; Reiser, G.; Reymann, K. Increase of prothrombin-mRNA after global cerebral ischemia in rats, with constant expression of protease nexin-1 and protease-activated receptors. Neurosci. Lett. 2002, 329, 181–184. [Google Scholar] [CrossRef]
  161. Thevenet, J.; Angelillo-Scherrer, A.; Price, M.; Hirt, L. Coagulation factor Xa activates thrombin in ischemic neural tissue. J. Neurochem. 2009, 111, 828–836. [Google Scholar] [CrossRef]
  162. Bushi, D.; Chapman, J.; Katzav, A.; Shavit-Stein, E.; Molshatzki, N.; Maggio, N.; Tanne, D. Quantitative detection of thrombin activity in an ischemic stroke model. J. Mol. Neurosci. 2013, 51, 844–850. [Google Scholar] [CrossRef] [PubMed]
  163. Hayosh, Z.e.I.; Bandora, E.A.; Shelestovich, N.; Nulman, M.; Bakon, M.; Yaniv, G.; Khaitovitch, B.; Balan, S.; Gerasimova, A.; Drori, T. In-thrombus thrombin secretion: A new diagnostic marker of atrial fibrillation in cryptogenic stroke. J. Neurointerv. Surg. 2020. [Google Scholar] [CrossRef]
  164. Vaughan, P.J.; Pike, C.J.; Cotman, C.W.; Cunningham, D.D. Thrombin receptor activation protects neurons and astrocytes from cell death produced by environmental insults. J. Neurosci. 1995, 15, 5389–5401. [Google Scholar] [CrossRef] [Green Version]
  165. Striggow, F.; Riek, M.; Breder, J.; Henrich-Noack, P.; Reymann, K.G.; Reiser, G. The protease thrombin is an endogenous mediator of hippocampal neuroprotection against ischemia at low concentrations but causes degeneration at high concentrations. Proc. Natl. Acad. Sci. USA 2000, 97, 2264–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Wu, X.; Zhang, W.; Li, J.Y.; Chai, B.X.; Peng, J.; Wang, H.; Mulholland, M.W. Induction of apoptosis by thrombin in the cultured neurons of dorsal motor nucleus of the vagus. Neurogastroenterol. Motil. 2011, 23, 279.e124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Hua, Y.; Wu, J.; Keep, R.; Hoff, J.; Xi, G. Thrombin exacerbates brain edema in focal cerebral ischemia. In Brain Edema XII; Springer: Berlin/Heidelberg, Germany, 2003; pp. 163–166. [Google Scholar]
  168. Hua, Y.; Wu, J.; Keep, R.F.; Nakamura, T.; Hoff, J.T.; Xi, G. Tumor necrosis factor-α increases in the brain after intracerebral hemorrhage and thrombin stimulation. Neurosurgery 2006, 58, 542–550. [Google Scholar] [CrossRef]
  169. Bodmer, D.; Vaughan, K.A.; Zacharia, B.E.; Hickman, Z.L.; Connolly, E.S. The molecular mechanisms that promote edema after intracerebral hemorrhage. Transl. Stroke Res. 2012, 3, 52–61. [Google Scholar] [CrossRef]
  170. Masasda, T.; Hua, Y.; Xi, G.; Yang, G.-Y.; Hoff, J.; Keep, R.; Nagao, S. Overexpression of interleukin-1 receptor antagonist reduces brain edema induced by intracerebral hemorrhage and thrombin. In Brain Edema XII; Springer: Berlin/Heidelberg, Germany, 2003; pp. 463–467. [Google Scholar]
  171. Nakamura, T.; Xi, G.; Park, J.-W.; Hua, Y.; Hoff, J.T.; Keep, R.F. Holo-transferrin and thrombin can interact to cause brain damage. Stroke 2005, 36, 348–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Hua, Y.; Keep, R.F.; Hoff, J.T.; Xi, G. Brain injury after intracerebral hemorrhage: The role of thrombin and iron. Stroke 2007, 38, 759–762. [Google Scholar] [CrossRef]
  173. Rashidian, J.; Iyirhiaro, G.; Aleyasin, H.; Rios, M.; Vincent, I.; Callaghan, S.; Bland, R.J.; Slack, R.S.; During, M.J.; Park, D.S. Multiple cyclin-dependent kinases signals are critical mediators of ischemia/hypoxic neuronal death in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2005, 102, 14080–14085. [Google Scholar] [CrossRef] [Green Version]
  174. Rao, H.V.; Thirumangalakudi, L.; Desmond, P.; Grammas, P. Cyclin D1, cdk4, and Bim are involved in thrombin-induced apoptosis in cultured cortical neurons. J. Neurochem. 2007, 101, 498–505. [Google Scholar] [CrossRef] [PubMed]
  175. Svedin, P.; Hagberg, H.; Sävman, K.; Zhu, C.; Mallard, C. Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia–ischemia. J. Neurosci. 2007, 27, 1511–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Boven, L.A.; Vergnolle, N.; Henry, S.D.; Silva, C.; Imai, Y.; Holden, J.; Warren, K.; Hollenberg, M.D.; Power, C. Up-regulation of proteinase-activated receptor 1 expression in astrocytes during HIV encephalitis. J. Immunol. 2003, 170, 2638–2646. [Google Scholar] [CrossRef]
  177. Ramos-Mandujano, G.; Vázquez-Juárez, E.; Hernández-Benítez, R.; Pasantes-Morales, H. Thrombin potently enhances swelling-sensitive glutamate efflux from cultured astrocytes. Glia 2007, 55, 917–925. [Google Scholar] [CrossRef]
  178. Ohnishi, M.; Katsuki, H.; Fujimoto, S.; Takagi, M.; Kume, T.; Akaike, A. Involvement of thrombin and mitogen-activated protein kinase pathways in hemorrhagic brain injury. Exp. Neurol. 2007, 206, 43–52. [Google Scholar] [CrossRef] [Green Version]
  179. Xi, G.; Keep, R.F.; Hua, Y.; Xiang, J.; Hoff, J.T. Attenuation of thrombin-induced brain edema by cerebral thrombin preconditioning. Stroke 1999, 30, 1247–1254. [Google Scholar] [CrossRef] [Green Version]
  180. Xi, G.; Keep, R.; Hua, Y.; Hoff, J. Thrombin preconditioning, heat shock proteins and thrombin-induced brain edema. In Brain Edema XI; Springer: Berlin/Heidelberg, Germany, 2000; pp. 511–515. [Google Scholar]
  181. Henrich-Noack, P.; Striggow, F.; Reiser, G.; Reymann, K.G. Preconditioning with thrombin can be protective or worsen damage after endothelin-1-induced focal ischemia in rats. J. Neurosci. Res. 2006, 83, 469–475. [Google Scholar] [CrossRef] [PubMed]
  182. Karabiyikoglu, M.; Hua, Y.; Keep, R.F.; Ennis, S.R.; Xi, G. Intracerebral hirudin injection attenuates ischemic damage and neurologic deficits without altering local cerebral blood flow. J. Cereb. Blood Flow Metab. 2004, 24, 159–166. [Google Scholar] [CrossRef] [PubMed]
  183. Yang, S.; Hua, Y.; Nakamura, T.; Keep, R.; Xi, G. Up-regulation of brain ceruloplasmin in thrombin preconditioning. In Brain Edema XIII; Springer: Berlin/Heidelberg, Germany, 2006; pp. 203–206. [Google Scholar]
  184. Wang, Y.; Luo, W.; Stricker, R.; Reiser, G. Protease-activated receptor-1 protects rat astrocytes from apoptotic cell death via JNK-mediated release of the chemokine GRO/CINC-1. J. Neurochem. 2006, 98, 1046–1060. [Google Scholar] [CrossRef]
  185. Hoffmann, M.-C.; Nitsch, C.; Scotti, A.; Reinhard, E.; Monard, D. The prolonged presence of glia-derived nexin, an endogenous protease inhibitor, in the hippocampus after ischemia-induced delayed neuronal death. Neuroscience 1992, 49, 397–408. [Google Scholar] [CrossRef]
  186. Cunningham, D.D.; Pulliam, L.; Vaughan, P.J. Protease nexin-1 and thrombin: Injury-related processes in the brain. Thromb. Haemost. 1993, 69, 168–171. [Google Scholar] [CrossRef]
  187. Executive Steering Committee on Behalf of the SPORTIF III Investigators. Stroke prevention with the oral direct thrombin inhibitor ximelagatran compared with warfarin in patients with non-valvular atrial fibrillation (SPORTIF III): Randomised controlled trial. Lancet 2003, 362, 1691–1698. [Google Scholar] [CrossRef]
  188. Diener, H.-C.; Sacco, R.L.; Easton, J.D.; Granger, C.B.; Bernstein, R.A.; Uchiyama, S.; Kreuzer, J.; Cronin, L.; Cotton, D.; Grauer, C. Dabigatran for prevention of stroke after embolic stroke of undetermined source. New Engl. J. Med. 2019, 380, 1906–1917. [Google Scholar] [CrossRef] [PubMed]
  189. Bai, Y.; Deng, H.; Shantsila, A.; Lip, G.Y. Rivaroxaban versus dabigatran or warfarin in real-world studies of stroke prevention in atrial fibrillation: Systematic review and meta-analysis. Stroke 2017, 48, 970–976. [Google Scholar] [CrossRef] [Green Version]
  190. Dans, A.L.; Connolly, S.J.; Wallentin, L.; Yang, S.; Nakamya, J.; Brueckmann, M.; Ezekowitz, M.; Oldgren, J.; Eikelboom, J.W.; Reilly, P.A. Concomitant use of antiplatelet therapy with dabigatran or warfarin in the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) trial. Circulation 2013, 127, 634–640. [Google Scholar] [CrossRef] [PubMed]
  191. Kim, K.S. Mechanisms of microbial traversal of the blood–brain barrier. Nat. Rev. Microbiol. 2008, 6, 625–634. [Google Scholar] [CrossRef] [Green Version]
  192. Kim, K.S. Microbial translocation of the blood–brain barrier. Int. J. Parasitol. 2006, 36, 607–614. [Google Scholar] [CrossRef]
  193. Mook-Kanamori, B.; Valls Seron, M.; Geldhoff, M.; Havik, S.; van der Ende, A.; Baas, F.; van der Poll, T.; Meijers, J.; Morgan, B.P.; Brouwer, M. Thrombin-activatable fibrinolysis inhibitor influences disease severity in humans and mice with pneumococcal meningitis. J. Thromb. Haemost. 2015, 13, 2076–2086. [Google Scholar] [CrossRef] [Green Version]
  194. Kremer Hovinga, J.; Franco, R.; Zago, M.; Ten Cate, H.; Westendorp, R.; Reitsma, P. A functional single nucleotide polymorphism in the thrombin-activatable fibrinolysis inhibitor (TAFI) gene associates with outcome of meningococcal disease. J. Thromb. Haemost. 2004, 2, 54–57. [Google Scholar] [CrossRef]
  195. Emonts, M.; De Bruijne, E.; Guimarães, A.; Declerck, P.; Leebeek, F.; De Maat, M.; Rijken, D.; Hazelzet, J.; Gils, A. Thrombin activatable fibrinolysis inhibitor is associated with severity and outcome of severe meningococcal infection in children. J. Thromb. Haemost. 2008, 6, 268–276. [Google Scholar] [CrossRef]
  196. Faust, S.N.; Levin, M.; Harrison, O.B.; Goldin, R.D.; Lockhart, M.S.; Kondaveeti, S.; Laszik, Z.; Esmon, C.T.; Heyderman, R.S. Dysfunction of endothelial protein C activation in severe meningococcal sepsis. New Engl. J. Med. 2001, 345, 408–416. [Google Scholar] [CrossRef] [PubMed]
  197. Baker, J.V.; Brummel-Ziedins, K.; Neuhaus, J.; Duprez, D.; Cummins, N.; Dalmau, D.; DeHovitz, J.; Lehmann, C.; Sullivan, A.; Woolley, I. HIV replication alters the composition of extrinsic pathway coagulation factors and increases thrombin generation. J. Am. Heart Assoc. 2013, 2, e000264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Hsue, P.Y.; Scherzer, R.; Grunfeld, C.; Nordstrom, S.M.; Schnell, A.; Kohl, L.P.; Nitta, E.; Martin, J.N.; Deeks, S.G.; Weiss, E.J. HIV infection is associated with decreased thrombin generation. Clin. Infect. Dis. 2012, 54, 1196–1203. [Google Scholar] [CrossRef] [Green Version]
  199. Ivey, N.S.; MacLean, A.G.; Lackner, A.A. Acquired immunodeficiency syndrome and the blood-brain barrier. J. Neurovirol. 2009, 15, 111–122. [Google Scholar] [CrossRef] [Green Version]
  200. Hurley, A.; Smith, M.; Karpova, T.; Hasley, R.B.; Belkina, N.; Shaw, S.; Balenga, N.; Druey, K.M.; Nickel, E.; Packard, B. Enhanced effector function of CD8+ T cells from healthy controls and HIV-infected patients occurs through thrombin activation of protease-activated receptor 1. J. Infect. Dis. 2013, 207, 638–650. [Google Scholar] [CrossRef] [Green Version]
  201. Noorbakhsh, F.; Vergnolle, N.; McArthur, J.C.; Silva, C.; Vodjgani, M.; Andrade-Gordon, P.; Hollenberg, M.D.; Power, C. Proteinase-activated receptor-2 induction by neuroinflammation prevents neuronal death during HIV infection. J. Immunol. 2005, 174, 7320–7329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Kim, W.; Zekas, E.; Lodge, R.; Susan-Resiga, D.; Marcinkiewicz, E.; Essalmani, R.; Mihara, K.; Ramachandran, R.; Asahchop, E.; Gelman, B. Neuroinflammation-induced interactions between protease-activated receptor 1 and proprotein convertases in HIV-associated neurocognitive disorder. Mol. Cell. Biol. 2015, 35, 3684–3700. [Google Scholar] [CrossRef] [Green Version]
  203. Avril, M.; Benjamin, M.; Dols, M.-M.; Smith, J.D. Interplay of Plasmodium falciparum and thrombin in brain endothelial barrier disruption. Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  204. Zeng, Y.; Liang, J.; Weng, C.; Lu, Z.; Zhou, Y. β-Arrestin 2 protects against neurological function defects in HSV-1-induced encephalitis mice. J. Med Virol. 2020, 92, 78–85. [Google Scholar] [CrossRef]
  205. Amlie-Lefond, C.; Kleinschmidt-Demasters, B.K.; Mahalingam, R.; Davis, L.E.; Gilden, D.H. The vasculopathy of varicella-zoster virus encephalitis. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 1995, 37, 784–790. [Google Scholar] [CrossRef] [PubMed]
  206. Itsekson-Hayosh, Z.; Shavit-Stein, E.; Katzav, A.; Rubovitch, V.; Maggio, N.; Chapman, J.; Harnof, S.; Pick, C.G. Minimal traumatic brain injury in mice: Protease-activated receptor 1 and thrombin-related changes. J. Neurotrauma 2016, 33, 1848–1854. [Google Scholar] [CrossRef]
  207. Ben Shimon, M.; Zeimer, T.; Shavit Stein, E.; Artan-Furman, A.; Harnof, S.; Chapman, J.; Eisenkraft, A.; Pick, C.G.; Maggio, N. Recovery from trauma induced amnesia correlates with normalization of thrombin activity in the mouse hippocampus. PLoS ONE 2017, 12, e0188524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Itzekson, Z.; Maggio, N.; Milman, A.; Shavit, E.; Pick, C.G.; Chapman, J. Reversal of trauma-induced amnesia in mice by a thrombin receptor antagonist. J. Mol. Neurosci. 2014, 53, 87–95. [Google Scholar] [CrossRef]
  209. Piao, C.-S.; Holloway, A.L.; Hong-Routson, S.; Wainwright, M.S. Depression following traumatic brain injury in mice is associated with down-regulation of hippocampal astrocyte glutamate transporters by thrombin. J. Cereb. Blood Flow Metab. 2019, 39, 58–73. [Google Scholar] [CrossRef] [PubMed]
  210. Tomkins, O.; Kaufer, D.; Korn, A.; Shelef, I.; Golan, H.; Reichenthal, E.; Soreq, H.; Friedman, A. Frequent blood–brain barrier disruption in the human cerebral cortex. Cell. Mol. Neurobiol. 2001, 21, 675–691. [Google Scholar] [CrossRef]
  211. Ballabh, P.; Braun, A.; Nedergaard, M. The blood–brain barrier: An overview: Structure, regulation, and clinical implications. Neurobiol. Dis. 2004, 16, 1–13. [Google Scholar] [CrossRef]
  212. Lee, K.R.; Drury, I.; Vitarbo, E.; Hoff, J.T. Seizures induced by intracerebral injection of thrombin: A model of intracerebral hemorrhage. J. Neurosurg. 1997, 87, 73–78. [Google Scholar] [CrossRef]
  213. Kelly, K.M. Thrombin: Is it on a par with seizures and epilepsy? Epilepsy Curr. 2008, 8, 110–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Chodobski, A.; Zink, B.J.; Szmydynger-Chodobska, J. Blood–brain barrier pathophysiology in traumatic brain injury. Transl. Stroke Res. 2011, 2, 492–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Pisapia, J.M.; Xu, X.; Kelly, J.; Yeung, J.; Carrion, G.; Tong, H.; Meghan, S.; El-Falaky, O.M.; Grady, M.S.; Smith, D.H. Microthrombosis after experimental subarachnoid hemorrhage: Time course and effect of red blood cell-bound thrombin-activated pro-urokinase and clazosentan. Exp. Neurol. 2012, 233, 357–363. [Google Scholar] [CrossRef] [Green Version]
  216. Stein, S.C.; Chen, X.-H.; Sinson, G.P.; Smith, D.H. Intravascular coagulation: A major secondary insult in nonfatal traumatic brain injury. J. Neurosurg. 2002, 97, 1373–1377. [Google Scholar] [CrossRef] [PubMed]
  217. Maggio, N.; Blatt, I.; Vlachos, A.; Tanne, D.; Chapman, J.; Segal, M. Treating seizures and epilepsy with anticoagulants? Front. Cell. Neurosci. 2013, 7, 19. [Google Scholar] [CrossRef] [Green Version]
  218. Isaeva, E.; Hernan, A.; Isaev, D.; Holmes, G.L. Thrombin facilitates seizures through activation of persistent sodium current. Ann. Neurol. 2012, 72, 192–198. [Google Scholar] [CrossRef] [Green Version]
  219. Lenz, M.; Shimon, M.B.; Benninger, F.; Neufeld, M.Y.; Shavit-Stein, E.; Vlachos, A.; Maggio, N. Systemic thrombin inhibition ameliorates seizures in a mouse model of pilocarpine-induced status epilepticus. J. Mol. Med. 2019, 97, 1567–1574. [Google Scholar] [CrossRef] [PubMed]
  220. Nierodzik, M.L.; Karpatkin, S. Thrombin induces tumor growth, metastasis, and angiogenesis: Evidence for a thrombin-regulated dormant tumor phenotype. Cancer Cell 2006, 10, 355–362. [Google Scholar] [CrossRef] [Green Version]
  221. Hu, L.; Lee, M.; Campbell, W.; Perez-Soler, R.; Karpatkin, S. Role of endogenous thrombin in tumor implantation, seeding, and spontaneous metastasis. Blood 2004, 104, 2746–2751. [Google Scholar] [CrossRef] [Green Version]
  222. Krenzlin, H.; Lorenz, V.; Alessandri, B. The involvement of thrombin in the pathogenesis of glioblastoma. J. Neurosci. Res. 2017, 95, 2080–2085. [Google Scholar] [CrossRef]
  223. Itsekson-Hayosh, Z.E.; Shavit-Stein, E.; Last, D.; Goez, D.; Daniels, D.; Bushi, D.; Gera, O.; Zibly, Z.; Mardor, Y.; Chapman, J. Thrombin activity and thrombin receptor in rat glioblastoma model: Possible markers and targets for intervention? J. Mol. Neurosci. 2015, 56, 644–651. [Google Scholar] [CrossRef] [PubMed]
  224. Kaufmann, R.; Zieger, M.; Tausch, S.; Henklein, P.; Nowak, G. Meizothrombin, an intermediate of prothrombin activation, stimulates human glioblastoma cells by interaction with PAR-1-type thrombin receptors. J. Neurosci. Res. 2000, 59, 643–648. [Google Scholar] [CrossRef]
  225. Auvergne, R.; Wu, C.; Connell, A.; Au, S.; Cornwell, A.; Osipovitch, M.; Benraiss, A.; Dangelmajer, S.; Guerrero-Cazares, H.; Quinones-Hinojosa, A. PAR1 inhibition suppresses the self-renewal and growth of A2B5-defined glioma progenitor cells and their derived gliomas in vivo. Oncogene 2016, 35, 3817–3828. [Google Scholar] [CrossRef] [Green Version]
  226. Kuhn, S.A.; Martin, M.; Brodhun, M.; Kratzsch, T.; Hanisch, U.-K.; Haberl, H. Overexpression of protease-activated receptor type 1 (PAR-1) in glioblastoma multiforme WHO IV cells and blood vessels revealed by NCAM-assisted glioblastoma border labeling. Neurol. Res. 2014, 36, 709–721. [Google Scholar] [CrossRef] [PubMed]
  227. de Almeida, V.H.; Monteiro, R.Q. Protease-activated receptor 1 (PAR1): A promising target for the treatment of glioblastoma. Transl. Cancer Res. 2016, 5, S1274–S1280. [Google Scholar] [CrossRef]
  228. Kaufmann, R.; Patt, S.; Kraft, R.; Zieger, M.; Henklein, P.; Neupert, G.; Nowak, G. PAR 1-type thrombin receptors are involved in thrombin-induced calcium signaling in human meningioma cells. J. Neuro Oncol. 1999, 42, 131–136. [Google Scholar] [CrossRef]
  229. Zieger, M.; Tausch, S.; Henklein, P.; Nowak, G.; Kaufmann, R. A novel PAR-1-type thrombin receptor signaling pathway: Cyclic AMP-independent activation of PKA in SNB-19 glioblastoma cells. Biochem. Biophys. Res. Commun. 2001, 282, 952–957. [Google Scholar] [CrossRef]
  230. Hayakawa, Y.; Kurimoto, M.; Nagai, S.; Kurosaki, K.; Tsuboi, Y.; Hamada, H.; Hayashi, N.; Endo, S. Thrombin-induced cell proliferation and platelet-derived growth factor-AB release from A172 human glioblastoma cells. J. Thromb. Haemost. 2007, 5, 2219–2226. [Google Scholar] [CrossRef]
  231. da Gama Fischer, J.D.S.; Carvalho, P.C.; da Costa Neves-Ferreira, A.G.; da Fonseca, C.O.; Perales, J.; da Costa Carvalho, M.D.G.; Domont, G.B. Anti-thrombin as a prognostic biomarker candidate for patients with recurrent glioblastoma multiform under treatment with perillyl alcohol. J. Exp. Oncol. 2008, 7, 285–290. [Google Scholar]
  232. Shavit-Stein, E.; Sheinberg, E.; Golderman, V.; Sharabi, S.; Wohl, A.; Gofrit, S.G.; Zivli, Z.; Shelestovich, N.; Last, D.; Guez, D. A novel compound targeting protease receptor 1 activators for the treatment of glioblastoma. Front. Neurol. 2018, 9, 1087. [Google Scholar] [CrossRef]
  233. Vianello, F.; Sambado, L.; Goss, A.; Fabris, F.; Prandoni, P. Dabigatran antagonizes growth, cell-cycle progression, migration, and endothelial tube formation induced by thrombin in breast and glioblastoma cell lines. Cancer Med. 2016, 5, 2886–2898. [Google Scholar] [CrossRef]
  234. Hua, Y.; Tang, L.; Keep, R.; Schallert, T.; Fewel, M.; Muraszko, K.; Hoff, J.; Xi, G. The role of thrombin in gliomas. J. Thromb. Haemost. 2005, 3, 1917–1923. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Relevance of thrombin to neurological disease and injury. The role of thrombin in epilepsy, CNS infections, mild traumatic brain injury, neurodegenerative diseases, stroke, and primary CNS tumors continues to be investigated.
Figure 1. Relevance of thrombin to neurological disease and injury. The role of thrombin in epilepsy, CNS infections, mild traumatic brain injury, neurodegenerative diseases, stroke, and primary CNS tumors continues to be investigated.
Biomolecules 11 00562 g001
Figure 2. Role of thrombin in cellular protection and apoptosis. At low concentrations, thrombin has anti-inflammatory effects and is involved in cellular protection and endothelial barrier stabilization. At high concentrations, thrombin leads to endothelial barrier disruption.
Figure 2. Role of thrombin in cellular protection and apoptosis. At low concentrations, thrombin has anti-inflammatory effects and is involved in cellular protection and endothelial barrier stabilization. At high concentrations, thrombin leads to endothelial barrier disruption.
Biomolecules 11 00562 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shlobin, N.A.; Har-Even, M.; Itsekson-Hayosh, Z.; Harnof, S.; Pick, C.G. Role of Thrombin in Central Nervous System Injury and Disease. Biomolecules 2021, 11, 562. https://doi.org/10.3390/biom11040562

AMA Style

Shlobin NA, Har-Even M, Itsekson-Hayosh Z, Harnof S, Pick CG. Role of Thrombin in Central Nervous System Injury and Disease. Biomolecules. 2021; 11(4):562. https://doi.org/10.3390/biom11040562

Chicago/Turabian Style

Shlobin, Nathan A., Meirav Har-Even, Ze’ev Itsekson-Hayosh, Sagi Harnof, and Chaim G. Pick. 2021. "Role of Thrombin in Central Nervous System Injury and Disease" Biomolecules 11, no. 4: 562. https://doi.org/10.3390/biom11040562

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop