Next Article in Journal
The Complex Interplay between Imbalanced Mitochondrial Dynamics and Metabolic Disorders in Type 2 Diabetes
Next Article in Special Issue
Retinal Injury Activates Complement Expression in Müller Cells Leading to Neuroinflammation and Photoreceptor Cell Death
Previous Article in Journal
Basal Ganglia Compensatory White Matter Changes on DTI in Alzheimer’s Disease
Previous Article in Special Issue
Astrocytosis, Inflammation, Axonal Damage and Myelin Impairment in the Internal Capsule following Striatal Ischemic Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Thrombin, a Key Driver of Pathological Inflammation in the Brain

1
Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
2
The Roskamp Institute, Sarasota, FL 34243, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(9), 1222; https://doi.org/10.3390/cells12091222
Submission received: 27 December 2022 / Revised: 21 February 2023 / Accepted: 20 April 2023 / Published: 23 April 2023

Abstract

:
Neurodegenerative diseases, including Alzheimer’s disease (AD), are major contributors to death and disability worldwide. A multitude of evidence suggests that neuroinflammation is critical in neurodegenerative disease processes. Exploring the key mediators of neuroinflammation in AD, a prototypical neurodegenerative disease, could help identify pathologic inflammatory mediators and mechanisms in other neurodegenerative diseases. Elevated levels of the multifunctional inflammatory protein thrombin are commonly found in conditions that increase AD risk, including diabetes, atherosclerosis, and traumatic brain injury. Thrombin, a main driver of the coagulation cascade, has been identified as important to pathological events in AD and other neurodegenerative diseases. Furthermore, recent evidence suggests that coagulation cascade-associated proteins act as drivers of inflammation in the AD brain, and studies in both human populations and animal models support the view that abnormalities in thrombin generation promote AD pathology. Thrombin drives neuroinflammation through its pro-inflammatory activation of microglia, astrocytes, and endothelial cells. Due to the wide-ranging pro-inflammatory effects of thrombin in the brain, inhibiting thrombin could be an effective strategy for interrupting the inflammatory cascade which contributes to neurodegenerative disease progression and, as such, may be a potential therapeutic target for AD and other neurodegenerative diseases.

1. Neuroinflammation: A Common Mechanism in Neurodegenerative Diseases

Neurodegenerative diseases are major contributors to death and disability worldwide. In 2016, neurological disorders were the leading cause of disability-adjusted life-years (DALYs; the sum of years of life lost) globally and the second leading cause of death [1,2]. These disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Multiple sclerosis (MS), and Amyotrophic lateral sclerosis (ALS), are pathological conditions defined by progressive and irreversible neuronal cell death and dysfunction [3]. While each of these diseases is characterized by unique patterns of neurodegeneration, as well as pathognomonic protein abnormalities which determine clinical presentation, neuroinflammation is an invariant feature of these disorders [4,5]. Exploring neuroinflammation in AD, a prototypical neurodegenerative disease, could help identify pathologic inflammatory mediators and mechanisms in other neurodegenerative diseases.
Pro-inflammatory cytokines and chemokines are significantly elevated in the brain of AD patients compared to healthy controls, as well as in animal models of AD [6,7,8,9]. These neuroinflammatory mediators appear early in the disease time course, suggesting their significant role in AD pathogenesis [8]. Animal studies show that chronic infusion of lipopolysaccharide (LPS) into the fourth ventricle of the rat brain reproduces the pathological changes found in the AD brain, supporting the role of neuroinflammation as a driver of AD pathology [10]. Additionally, a robust literature demonstrates the activation of inflammatory processes in brain areas closely associated with AD pathology, and activated microglia, thought to be key drivers of neuroinflammation, are found in or near the pathologic lesions of AD [11,12,13]. Circulating levels of systemic inflammatory mediators, such as interleukin (IL)-1β and tumor necrosis factor (TNFα), are chronically upregulated in AD, while an increased risk for AD is linked to systemic inflammatory conditions such as Type 2 diabetes, obesity, and rheumatoid arthritis [14,15]. Additionally, large-scale genome-wide association studies highlight brain-associated genetic variants linked to neuroinflammation in AD, including TREM2, CD33, and CR1 [16,17,18,19]. The idea that inflammatory mediators are linked to cognitive impairment is reinforced by experiments in apolipoprotein E (ApoE) knockout mice, which show induction of IL-6 and IL-8 in the brain microcirculation is associated with impaired cognition [20]. The link between neurovascular inflammation and cognitive function is further supported by data showing that reducing the expression of inflammation proteins in the cerebral microcirculation improves cognition in animal models of AD [21].
There is a multitude of experimental evidence suggesting that neuroinflammation acts as a critical pathological event, initiating and driving neurodegenerative processes associated with many neurological diseases. Therefore, identifying the key mediators of pathological inflammation in the brain is important to understanding and treating these devastating conditions. Elevated levels of the multifunctional inflammatory protein thrombin are commonly found in conditions that increase AD risk, including diabetes, atherosclerosis, and traumatic brain injury (TBI) [22,23,24,25]. Even though thrombin has been implicated in AD since the 1990s, when it was first detected in senile plaques and neurofibrillary tangles [26], the relevance of thrombin in AD pathology, and other neurodegenerative diseases, has not been widely explored.

2. Thrombin, a Key Driver of Neuroinflammation

Brain endothelial cells in AD express elevated levels of proteins that mediate cell adhesion and migration of inflammatory cells, including intracellular adhesion molecule (ICAM)-1, monocyte chemoattractant protein (MCP)-1, and CAP37 [27,28,29]. In addition, brain microvessels isolated from AD patient samples release higher levels of inflammatory mediators such as thrombin, nitric oxide (NO), TNFα, IL-1β, IL-6, IL-8, and matrix metalloproteinases (MMPs), compared to brain microvessels isolated from age-matched healthy controls [27,30,31]. The extensive number of chemokines and cytokines that are over-expressed in the vasculature in AD are consistent with an “activated proinflammatory endothelial phenotype” [32]. The multifunctional serine protease thrombin is a primary driver of endothelial activation and inflammation in both the periphery and the CNS in pathological conditions [33]. This pluripotent inflammatory mediator has a diverse array of cellular targets throughout the body and CNS, including endothelial cells, monocytes, neurons, astrocytes, pericytes, and microglia, through its activation of the protease-activated receptors (PARs) [34,35]. The importance of thrombin as a modulator of pathological inflammation in the brain is supported by data demonstrating that elevated levels of inflammatory cytokines evoked by systemic administration of LPS are decreased by inhibiting thrombin [36]. Furthermore, a direct role for thrombin in the inflammatory response characteristic of several neurodegenerative diseases has been suggested. PAR expression is increased in the substantia nigra of PD patients, and this increase is associated with the loss of dopaminergic cells, neuroinflammation, and oxidative stress [37,38,39]. In an animal model of MS, experimental autoimmune encephalomyelitis (EAE), elevated thrombin activity precedes the onset of neurological signs. Furthermore, thrombin activity was found to increase at the disease peak and was correlated with microglial activation, demyelination, axonal damage, and clinical severity [40]. In the same regard, expression of thrombin and PARs is increased in the AD brain [41,42].
The importance of thrombin as a regulator of cerebrovascular inflammation is further suggested by data in AD mice showing that thrombin inhibition causes a reduction in the production of inflammatory proteins. Furthermore, inflammation caused by hypoxia in brain endothelial cells is diminished by inhibiting thrombin [21]. These data showing the ability of thrombin to regulate brain vascular inflammation are important, as cerebral microcirculation in AD has been shown to produce thrombin. In this regard, thrombin message and protein levels are significantly increased in brain microvessels from AD patients and AD transgenic mice compared to controls [30,43]. In addition, immunoreactivity for the major thrombin inhibitor protease nexin 1 (PN1) is significantly decreased around blood vessels in AD, further suggesting that brain blood vessels are releasing thrombin in AD [44,45].
Finally, thrombin is widely appreciated as a central regulator of the coagulation cascade [46]. The coagulation cascade, consisting of both intrinsic and extrinsic pathways, involves sequential activation of clotting factors culminating ultimately in hemostasis. At the nexus of both intrinsic and extrinsic pathways is thrombin [47]. Thrombin cleaves fibrinogen, a soluble protein, into insoluble fibrin. Thrombin, through activation of Factor XIII, also affects the cross-linking of fibrin monomers to produce a firm fibrin clot and prevents clot fibrinolysis via activation of Factor XI [48]. Bidirectional communication between coagulation and inflammation is critical to the physiologic response to vascular injury [49]. Inflammation activates coagulation as a means of containing inflammatory damage, and coagulation-related factors, including thrombin, tissue factor (TF), and fibrin, can influence the extent and nature of inflammation [49,50]. For example, TF is pro-inflammatory because it can activate the PARs [51] and cause an increase in a variety of inflammatory proteins, including cytokines, chemokines, and adhesion proteins. Similarly, fibrin has been shown to regulate expression of inflammatory cytokines and chemokines and reactive oxygen species (ROS) in the brain [52,53,54]. Fibrin’s pro-inflammatory effects result in the activation of glia and disruption of blood–brain barrier (BBB) function [55]. Both fibrinogen and fibrin induce leukocyte migration and directly modulate the inflammatory response of leukocytes and endothelial cells [56].
Accumulating evidence demonstrates that coagulation cascade-associated proteins can initiate or drive inflammation in neurodegenerative diseases [33,38,52,57,58,59]. A role for the coagulation cascade in the development of MS pathology is supported by the finding that coagulation proteins are present in chronic active plaques in MS patients [60]. Similarly, in AD and AD animal models, fibrinogen co-localizes with amyloid plaques, pericyte loss, dystrophic neurites, and activated microglia [55,61]. The importance of this coagulation protein in the pathogenesis of AD is further suggested by data in AD mice, showing that fibrinogen depletion reduces cognitive dysfunction and AD-related pathology. It is important to note that thrombin is critical for fibrin formation, and therefore thrombin may function as an upstream mediator of the inflammatory effects of fibrin in the AD brain [46].

3. Thrombin-Mediated Inflammation: A Shared Mechanism among AD Risk Factors

Several diseases and conditions have been documented to increase the risk of developing AD. Among the most well-documented modifiable risk factors are atherosclerosis, diabetes, and TBI. A common thread among these risk factors is elevated levels of the pro-inflammatory protein thrombin.

3.1. Atherosclerosis

Given thrombin’s multiple effects on endothelial cells, which result in increased immune cell adhesion and inflammatory protein release, it is not surprising that thrombin is a driver of vascular pathology in atherosclerosis. Thrombin’s ability to promote atherosclerotic plaque formation reflects its key role as both a mediator of clot formation and inflammation [23,49,62]. Activation of PAR-1 by thrombin initiates signaling cascades in the vessel wall cell types, including endothelial cells, smooth muscle cells, and macrophages that are participatory in plaque genesis and development. In addition, the release of procoagulant factors, such as TF, which are evoked by thrombin, contributes to endothelial injury and plaque expansion [63].
Thrombin and its receptors (PARs) are elevated in atherosclerosis and are found around atherosclerotic plaques [64]. Thrombin is involved in the recruitment of circulating monocytes to atherosclerotic plaques by increasing expression of MCP-1, ICAM-1, vascular cell adhesion molecule (VCAM)-1, and platelet-derived growth factor (PDGF) [23,65,66,67], and increases in thrombin are associated with increased inflammation, angiogenesis, and cell proliferation in atherosclerotic models [23,68]. In addition, thrombin has pro-inflammatory effects on the vessel wall through its effects on NADPH oxidase and the NF-kB pathway. This activation of the NF-kB pathway leads to a number of downstream effects, including increased cell infiltration, increased expression of inflammatory molecules, promotion of hypercoagulability, and accelerated plaque development [69]. The observation of hypercoagulability in early atherosclerotic lesions supports the idea that the thrombin-stimulated release of procoagulant enzymes plays a fundamental role in the atherogenic process. Finally, genetic manipulations which increase thrombin exacerbate atherosclerosis-related pathology [70], while treatment with thrombin inhibitors reduces atherosclerosis-related pathology in animal models [71,72,73,74]. For example, thrombin inhibition attenuated atherosclerotic plaque formation and reduced both inflammation and PAR-1 expression in an ApoE-deficient mouse model of atherosclerosis [71].

3.2. Diabetes

Several lines of evidence support a role for thrombin in diabetes. Diabetic patients have increased thrombin levels in their blood, and high thrombin levels have been linked to poor diabetic control [75]. Hyperglycemia enhances thrombin generation leading to a hypercoagulable state in Type 2 diabetes [75,76,77]. Total thrombin generation and platelet reactivity are increased in Type 2 diabetes and older obese women [78]. An animal study examining the effects of obesity and metabolic syndrome, conditions frequently observed in diabetes, showed increased thrombin generation as early as 25 weeks of age [79]. Increased thrombin generation was also demonstrable in 7 to 10-week-old diabetic Zucker rats [80]. A study in the STZ rat demonstrated the importance of the thrombin pathway in a rat diabetic neuropathy model. Here, elevated thrombin activity was observed in the diabetic sciatic nerve correlating with the destruction of nodal histology and altered electrophysiological nerve conduction. Treatment with thrombin inhibitors ameliorated structural and electrophysiological deficits in diabetic animals [81].
Thrombin receptors are increased in the vasculature in the Type 1 diabetic animal model, streptozotocin (STZ) [82]. Thrombin, through its pro-inflammatory effects on vascular endothelial cells, is linked to endothelial dysfunction in diabetes [83]. In this regard, thrombin has been shown to increase oxidative stress in endothelial cells in diabetes via calcium-mediated intracellular pathways that regulate the transcription factor KLF14 and PLK1 kinase pathways [84]. In vitro, data support a direct role for thrombin in the noxious effects of glucose on endothelial cells. Treatment of brain endothelial cells with high glucose causes increased expression of inflammatory proteins (TNFα, IL-6, MMP-2, MMP-9) and ROS, an effect that is mitigated by inhibiting thrombin [85].

3.3. Traumatic Brain Injury (TBI)

Thrombin has been implicated in the pathological effects of TBI. Studies have identified elevated thrombin levels following TBI in both human and animal models [86,87,88,89,90], and it has been reported that neurons are exposed to elevated levels of thrombin following TBI [91,92]. A recent study found that thrombin levels rise in the first-hour post-trauma and again after 72 h [25]. Thrombin signaling activity after TBI has been associated with negative cognitive outcomes, including amnesia [93]. An increase in depressive behavior following TBI was also associated with thrombin-mediated down-regulation of hippocampal astrocyte glutamate transporters [94]. Thrombin’s negative effects after TBI have been linked to thrombin activation of both PAR-1 and PAR-4 [25,95].
Thrombin has been specifically implicated in inflammatory processes after TBI. Increases in thrombin are associated with the upregulation of adhesion molecules, including ICAM-1, by the cerebrovasculature, which is associated with increased peripheral immune cell migration into the brain [96,97,98]. Furthermore, elevated thrombin in TBI is associated with astrocyte activation and astrocyte-mediated inflammation [88], and inhibition of thrombin signaling after TBI via PAR-4 antagonism reduces expression of inflammatory genes related to the NF-κB signaling pathway [95].

4. Cellular Effects of Thrombin in the Brain: Implications for AD

Along with inflammation, thrombin has been associated with the classical pathological hallmarks of AD. Thrombin can trigger both tau and Aβ accumulation. Persistent thrombin signaling induces tau aggregation and hippocampal degeneration [99,100]. Thrombin induces secretion of AβPP, while Aβ promotes thrombin generation through Factor XII-mediated Factor XI activation [101]. Furthermore, thrombin delivered directly into the brain via intracerebroventricular injection results in significant neuropathology, enlargement of cerebral ventricles, increased TUNEL-positive cells, astrogliosis, ApoE fragments, and cognitive impairments [102,103]. Thrombin regulates the brain’s inflammatory response via pro-inflammatory activation of several cell types, including microglia, astrocytes, and especially endothelial cells of the BBB.

4.1. Microglia

Thrombin has been found to activate a pro-inflammatory state in microglia. The microglial response to thrombin includes several processes that modulate or contribute to microglia activation and/or apoptosis. Exposure of the microglial cell line BV2 to thrombin induces a pro-inflammatory response, including IL-1β release [104] and an increase in ROS and activation of the NLRP3 inflammasome [105], a component of the innate immune system. Primary microglia also take on a pro-inflammatory phenotype when stimulated by thrombin, characterized by increased production of cytokines, ROS, and NO [105,106,107,108]. Thrombin’s pro-inflammatory effects in microglia are directly linked to PAR-1 activation, which leads to the upregulation of microglial CD40 and increased TNFα production [109]. Additionally, thrombin activation of a TNFα/TNFR-dependent pathway downregulates expression of the mRNA species miR181c, which then promotes NF-κB target gene expression and related activity [110].

4.2. Astrocytes

Similar to microglia, astrocytes exhibit a shift towards a pro-inflammatory phenotype in response to thrombin. Thrombin induces MMP-9 expression and promotes cell migration via activation of the c-Src/Jak2/PDGFR/PI3K/Akt/PKCδ pathway in rat brain astrocytes [111]. Thrombin exposure can also alter astrocytic function, marked by disruptions in glutamate transport and altered stellation [88,112]. Furthermore, thrombin treatment in vivo increases GFAP expression in the rat hippocampus, indicating a likely increase in pro-inflammatory activation of astrocytes [103].

4.3. Endothelial Cells

Thrombin exerts many pro-inflammatory effects on endothelial cells. Thrombin modulates its effects on cerebrovascular endothelial cells via altered gene expression of pro-inflammatory mediators. Activation of brain endothelial cells by thrombin leads to increased expression and/or release of pro-inflammatory proteins, including ROS, NO, ICAM-1, VCAM-1, MCP-1, TNFα, IL-1, IL-6, and IL-8, as well as thrombin itself [97,98]. Additionally, thrombin stimulation modifies the organization of cell-to-cell junctions between endothelial cells and cytoskeletal actin filaments, leading to increased BBB permeability [98,113,114]. Finally, thrombin at the BBB is particularly problematic because brain endothelial cells can both release thrombin and respond to it via PAR-1 and PAR-3 [30,98]. Thus, thrombin can not only activate neighboring cells, but it can also act in an autocrine manner on the endothelium stimulating a noxious feed-forward cycle (Figure 1).

5. Thrombin: A Novel Inflammatory Therapeutic Target in AD

Since neuroinflammation contributes to the progression of AD and thrombin has wide-ranging pro-inflammatory effects in the brain, inhibiting thrombin could be an effective strategy for interrupting the inflammatory cascade which contributes to disease progression in AD.
Studies using anticoagulant therapies in both human patients and animal models support the idea that abnormalities in thrombin generation may promote AD-related pathology. In line with this notion, an open-label study of a hirudin (natural antithrombin anticoagulant) compound in eighty-four patients with mild-to-moderate AD found that hirudin plus donepezil reduced the rate of cognitive decline compared to donepezil alone [115]. Additionally, in a retrospective cohort study, the use of direct-acting anticoagulants, including dabigatran, was associated with a lower risk of dementia [116]. Although a placebo-controlled double-blind clinical trial of dabigatran in AD has yet to be performed, the possibility that anticoagulant therapy, specifically targeting thrombin, could be useful in AD is supported by a number of population-based studies that show reduced incidence of dementia in atrial fibrillation patients taking direct oral anticoagulants that target thrombin [116,117,118,119].
In transgenic AD mouse models, targeting thrombin has been shown to reduce brain inflammation as well as improve AD pathology. Treatment of transgenic AD mice with low molecular weight heparin enoxaparin reduces plaques and Aβ accumulation and improves spatial memory [120]. Treatment of 3xTgAD mice with the thrombin inhibitor dabigatran decreases cerebrovascular expression of inflammatory proteins, including thrombin, IL-6, MCP-1, and MMPs [21]. Long-term (1 year) dabigatran treatment of the TgCRND8 AD mouse model reduces neuroinflammation, amyloid plaques, and amyloid oligomers and preserves cognitive function [121]. Finally, treatment of a tau-based AD model (rTg4510) with a thrombin inhibitor decreases inflammation and oxidative stress-related and AD marker proteins (ratio of phospho-tau to total tau) [122]. Taken together, these data support the idea that inhibiting thrombin in the AD brain reduces inflammation and improves AD pathology and, as such, could be useful as part of a therapeutic regimen.
It should be noted that the use of any anticoagulant brings up safety concerns about the possibility of unwanted bleeding. Safety issues regarding the use of direct oral anticoagulants (DOACs), which include the direct thrombin inhibitor dabigatran, as well as Factor Xa inhibitors apixaban, edoxaban and rivaroxaban, have been extensively explored [116,123,124,125,126,127,128,129,130]. Treatment with DOACs was associated with lower rates of both stroke and systemic embolism when compared with warfarin [131]. In addition, both apixaban and dabigatran exhibited reduced rates of major bleeding, including gastrointestinal bleeding and intracranial hemorrhage, compared with warfarin [132]. A study in patients with an average age of 71.4 ± 8.6 years old showed that the use of dabigatran for over 30 months had minimal side effects [125].
A comparison of dabigatran and warfarin showed that dabigatran reduced the incidence of intracranial hemorrhage by 66% [132]. Dabigatran also exhibited an improved safety pattern than the Factor Xa-targeting rivaroxaban [133]. Rivaroxaban use was found to increase intracranial and extracranial hemorrhage compared to dabigatran [133]. The increased risk of bleeding with rivaroxaban compared to dabigatran may reflect the extent of blood–brain barrier penetration. Pharmacokinetic analysis of DOACs indicates that the greatest risk for adverse events is related to the level of BBB penetration. Evaluating the physicochemical and pharmacologic properties of DOACs reveals that BBB penetration is greatest with rivaroxaban, followed by apixaban, with dabigatran having the lowest potential risk for BBB penetration [134]. Finally, in mouse models of AD and cerebral amyloid angiopathy (CAA), treatment with dabigatran was not linked with increased intracerebral hemorrhage or microbleeds [135,136].

6. Conclusions

Drugs that provide symptomatic benefits for the treatment of AD, such as cholinesterase inhibitors and the glutamate antagonist memantine, have been available for decades [137]. However, developing effective therapeutic strategies for mitigating the progression of AD has proved challenging [138]. The pathogenesis of late-onset AD is complex and multifactorial; no single mechanism or pathologic mediator can account for AD progression. It is, therefore, not surprising that single-targeted amyloid-β-directed therapies have not shown significant clinical benefit to date. Recently, aducanumab, a monoclonal anti-Aβ oligomer antibody, received accelerated approval from FDA based on its ability to lower brain amyloid levels, although clinical benefit has yet to be determined [139], while late-stage lecanemab, a monoclonal antibody targeting Aβ aggregation, did appear to slow cognitive decline in people with early disease in a Phase 3 trial, but the effect was modest [140]. These data reinforce the idea that single-target therapies are not effective in mitigating the complex pathophysiology of AD.
Inflammation has long been appreciated as a central mechanism contributing to disease progression in AD. Therefore, targeting inflammation is a rational approach to AD prevention and therapy [141]. In this regard, long-term non-steroidal anti-inflammatory drug (NSAID) treatment is also associated with both reduced Aβ deposition in mouse models of AD and reduced number of plaque-associated microglia [142]. Retrospective epidemiological studies in humans suggest that a wide variety of NSAIDs may significantly reduce one’s lifetime risk of developing AD [143,144,145,146]. A recent meta-analysis of sixteen studies demonstrates that present or previous use of NSAIDs decreases the relative risk of AD [147]. However, despite these epidemiological data, anti-inflammatory placebo-controlled trials have yielded negative results.
A growing consensus in the AD treatment landscape argues for an innovative approach to AD therapy [148]. Following the example of other diseases such as cancer and HIV, parallel administration of two drugs that target different pathways could be employed, as well as using repurposed drugs as add-on treatments to existing standard-of-care protocols. Targeting the multifunctional inflammatory protein thrombin in conjunction with therapies directed at other AD targets (amyloid, tau) could be an innovative and novel therapeutic strategy in the fight against this devastating disease.

Author Contributions

Conceptualization, J.I. and P.G.; investigation, J.I. and P.G.; resources, J.I.; writing—original draft preparation, J.I. and P.G.; writing—review and editing, J.I. and P.G.; supervision, P.G.; project administration, J.I.; funding acquisition, J.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Department of Defense (DoD), grant number W81XWH2210280.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Figure created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Feigin, V.L.; Nichols, E.; Alam, T.; Bannick, M.S.; Beghi, E.; Blake, N.; Culpepper, W.J.; Dorsey, E.R.; Elbaz, A.; Ellenbogen, R.G.; et al. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef] [PubMed]
  2. Feigin, V.L.; Vos, T.; Alahdab, F.; Amit, A.M.L.; Bärnighausen, T.W.; Beghi, E.; Beheshti, M.; Chavan, P.P.; Criqui, M.H.; Desai, R.; et al. Burden of Neurological Disorders across the US from 1990–2017: A Global Burden of Disease Study. JAMA Neurol. 2021, 78, 165–176. [Google Scholar] [CrossRef] [PubMed]
  3. Kovacs, G.G. Concepts and classification of neurodegenerative diseases. Handb. Clin. Neurol. 2017, 145, 301–307. [Google Scholar] [CrossRef] [PubMed]
  4. Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef]
  5. Chitnis, T.; Weiner, H.L. CNS inflammation and neurodegeneration. J. Clin. Investig. 2017, 127, 3577–3587. [Google Scholar] [CrossRef]
  6. Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G.M.; Cooper, N.R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B.L.; et al. Inflammation and Alzheimer’s disease. Neurobiol. Aging 2000, 21, 383–421. [Google Scholar] [CrossRef]
  7. Akiyama, H. Inflammatory response in Alzheimer’s disease. Tohoku J. Exp. Med. 1994, 174, 295–303. [Google Scholar] [CrossRef]
  8. Calsolaro, V.; Edison, P. Neuroinflammation in Alzheimer’s disease: Current evidence and future directions. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2016, 12, 719–732. [Google Scholar] [CrossRef]
  9. Cartier, L.; Hartley, O.; Dubois-Dauphin, M.; Krause, K.H. Chemokine receptors in the central nervous system: Role in brain inflammation and neurodegenerative diseases. Brain Res. Rev. 2005, 48, 16–42. [Google Scholar] [CrossRef]
  10. Hauss-Wegrzyniak, B.; Dobrzanski, P.; Stoehr, J.D.; Wenk, G.L. Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res. 1998, 780, 294–303. [Google Scholar] [CrossRef]
  11. Matsuoka, Y.; Picciano, M.; Malester, B.; LaFrancois, J.; Zehr, C.; Daeschner, J.M.; Olschowka, J.A.; Fonseca, M.I.; O’Banion, M.K.; Tenner, A.J.; et al. Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer’s disease. Am. J. Pathol. 2001, 158, 1345–1354. [Google Scholar] [CrossRef] [PubMed]
  12. Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
  13. Fakhoury, M. Microglia and Astrocytes in Alzheimer’s Disease: Implications for Therapy. Curr. Neuropharmacol. 2018, 16, 508–518. [Google Scholar] [CrossRef] [PubMed]
  14. Lutshumba, J.; Nikolajczyk, B.S.; Bachstetter, A.D. Dysregulation of Systemic Immunity in Aging and Dementia. Front. Cell. Neurosci. 2021, 15, 652111. [Google Scholar] [CrossRef] [PubMed]
  15. Newcombe, E.A.; Camats-Perna, J.; Silva, M.L.; Valmas, N.; Huat, T.J.; Medeiros, R. Inflammation: The link between comorbidities, genetics, and Alzheimer’s disease. J. Neuroinflamm. 2018, 15, 276. [Google Scholar] [CrossRef]
  16. Jones, L.; Holmans, P.A.; Hamshere, M.L.; Harold, D.; Moskvina, V.; Ivanov, D.; Pocklington, A.; Abraham, R.; Hollingworth, P.; Sims, R.; et al. Genetic evidence implicates the immune system and cholesterol metabolism in the aetiology of Alzheimer’s disease. PLoS ONE 2010, 5, e13950. [Google Scholar] [CrossRef] [PubMed]
  17. Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.; Younkin, S.; et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 117–127. [Google Scholar] [CrossRef]
  18. Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.; Jonsson, P.V.; Snaedal, J.; Bjornsson, S.; Huttenlocher, J.; Levey, A.I.; Lah, J.J.; et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 107–116. [Google Scholar] [CrossRef]
  19. Lambert, J.C.; Ibrahim-Verbaas, C.A.; Harold, D.; Naj, A.C.; Sims, R.; Bellenguez, C.; DeStafano, A.L.; Bis, J.C.; Beecham, G.W.; Grenier-Boley, B.; et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 2013, 45, 1452–1458. [Google Scholar] [CrossRef]
  20. Evola, M.; Hall, A.; Wall, T.; Young, A.; Grammas, P. Oxidative stress impairs learning and memory in apoE knockout mice. Pharmacol. Biochem. Behav. 2010, 96, 181–186. [Google Scholar] [CrossRef]
  21. 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] [PubMed]
  22. Jaberi, N.; Soleimani, A.; Pashirzad, M.; Abdeahad, H.; Mohammadi, F.; Khoshakhlagh, M.; Khazaei, M.; Ferns, G.A.; Avan, A.; Hassanian, S.M. Role of thrombin in the pathogenesis of atherosclerosis. J. Cell. Biochem. 2019, 120, 4757–4765. [Google Scholar] [CrossRef] [PubMed]
  23. Kalz, J.; ten Cate, H.; Spronk, H.M. Thrombin generation and atherosclerosis. J. Thromb. Thrombolysis 2014, 37, 45–55. [Google Scholar] [CrossRef] [PubMed]
  24. Ay, L.; Hoellerl, F.; Ay, C.; Brix, J.M.; Koder, S.; Schernthaner, G.H.; Pabinger, I.; Schernthaner, G. Thrombin generation in type 2 diabetes with albuminuria and macrovascular disease. Eur. J. Clin. Investig. 2012, 42, 470–477. [Google Scholar] [CrossRef]
  25. 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]
  26. 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] [PubMed]
  27. Grammas, P.; Ovase, R. Inflammatory factors are elevated in brain microvessels in Alzheimer’s disease. Neurobiol. Aging 2001, 22, 837–842. [Google Scholar] [CrossRef]
  28. Frohman, E.M.; Frohman, T.C.; Gupta, S.; de Fougerolles, A.; van den Noort, S. Expression of intercellular adhesion molecule 1 (ICAM-1) in Alzheimer’s disease. J. Neurol. Sci. 1991, 106, 105–111. [Google Scholar] [CrossRef] [PubMed]
  29. Pereira, H.A.; Kumar, P.; Grammas, P. Expression of CAP37, a novel inflammatory mediator, in Alzheimer’s disease. Neurobiol. Aging 1996, 17, 753–759. [Google Scholar] [CrossRef]
  30. Yin, X.; Wright, J.; Wall, T.; Grammas, P. Brain endothelial cells synthesize neurotoxic thrombin in Alzheimer’s disease. Am. J. Pathol. 2010, 176, 1600–1606. [Google Scholar] [CrossRef]
  31. Grammas, P.; Tripathy, D.; Sanchez, A.; Yin, X.; Luo, J. Brain microvasculature and hypoxia-related proteins in Alzheimer’s disease. Int. J. Clin. Exp. Pathol. 2011, 4, 616–627. [Google Scholar] [PubMed]
  32. Grammas, P. Neurovascular dysfunction, inflammation and endothelial activation: Implications for the pathogenesis of Alzheimer’s disease. J. Neuroinflamm. 2011, 8, 26. [Google Scholar] [CrossRef] [PubMed]
  33. 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]
  34. Coughlin, S.R. Protease-activated receptors in hemostasis, thrombosis and vascular biology. J. Thromb. Haemost. 2005, 3, 1800–1814. [Google Scholar] [CrossRef]
  35. Coughlin, S.R. Thrombin signalling and protease-activated receptors. Nature 2000, 407, 258–264. [Google Scholar] [CrossRef] [PubMed]
  36. Shavit Stein, E.; Ben Shimon, M.; Artan Furman, A.; Golderman, V.; Chapman, J.; Maggio, N. Thrombin Inhibition Reduces the Expression of Brain Inflammation Markers upon Systemic LPS Treatment. Neural Plast. 2018, 2018, 7692182. [Google Scholar] [CrossRef]
  37. Luo, W.; Wang, Y.; Reiser, G. Protease-activated receptors in the brain: Receptor expression, activation, and functions in neurodegeneration and neuroprotection. Brain Res. Rev. 2007, 56, 331–345. [Google Scholar] [CrossRef]
  38. 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]
  39. 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]
  40. 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.; et al. Early detection of thrombin activity in neuroinflammatory disease. Ann. Neurol. 2014, 75, 303–308. [Google Scholar] [CrossRef]
  41. 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] [PubMed]
  42. 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] [PubMed]
  43. 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]
  44. 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]
  45. Wagner, S.L.; Geddes, J.W.; Cotman, C.W.; Lau, A.L.; Gurwitz, D.; Isackson, P.J.; Cunningham, D.D. Protease nexin-1, an antithrombin with neurite outgrowth activity, is reduced in Alzheimer disease. Proc. Natl. Acad. Sci. USA 1989, 86, 8284–8288. [Google Scholar] [CrossRef]
  46. Palta, S.; Saroa, R.; Palta, A. Overview of the coagulation system. Indian J. Anaesth. 2014, 58, 515–523. [Google Scholar] [CrossRef]
  47. Mann, K.G.; Butenas, S.; Brummel, K. The dynamics of thrombin formation. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 17–25. [Google Scholar] [CrossRef] [PubMed]
  48. Narayanan, S. Multifunctional roles of thrombin. Ann. Clin. Lab. Sci. 1999, 29, 275–280. [Google Scholar]
  49. Esmon, C.T. Crosstalk between inflammation and thrombosis. Maturitas 2008, 61, 122–131. [Google Scholar] [CrossRef]
  50. Levi, M.; van der Poll, T. Two-way interactions between inflammation and coagulation. Trends Cardiovasc. Med. 2005, 15, 254–259. [Google Scholar] [CrossRef]
  51. Witkowski, M.; Landmesser, U.; Rauch, U. Tissue factor as a link between inflammation and coagulation. Trends Cardiovasc. Med. 2016, 26, 297–303. [Google Scholar] [CrossRef] [PubMed]
  52. Petersen, M.A.; Ryu, J.K.; Akassoglou, K. Fibrinogen in neurological diseases: Mechanisms, imaging and therapeutics. Nat. Rev. Neurosci. 2018, 19, 283–301. [Google Scholar] [CrossRef] [PubMed]
  53. Davalos, D.; Akassoglou, K. Fibrinogen as a key regulator of inflammation in disease. Semin. Immunopathol. 2012, 34, 43–62. [Google Scholar] [CrossRef]
  54. Cortes-Canteli, M.; Zamolodchikov, D.; Ahn, H.J.; Strickland, S.; Norris, E.H. Fibrinogen and altered hemostasis in Alzheimer’s disease. J. Alzheimer’s Dis. 2012, 32, 599–608. [Google Scholar] [CrossRef] [PubMed]
  55. Merlini, M.; Rafalski, V.A.; Rios Coronado, P.E.; Gill, T.M.; Ellisman, M.; Muthukumar, G.; Subramanian, K.S.; Ryu, J.K.; Syme, C.A.; Davalos, D.; et al. Fibrinogen Induces Microglia-Mediated Spine Elimination and Cognitive Impairment in an Alzheimer’s Disease Model. Neuron 2019, 101, 1099–1108.e1096. [Google Scholar] [CrossRef] [PubMed]
  56. Luyendyk, J.P.; Schoenecker, J.G.; Flick, M.J. The multifaceted role of fibrinogen in tissue injury and inflammation. Blood 2019, 133, 511–520. [Google Scholar] [CrossRef] [PubMed]
  57. Davalos, D.; Ryu, J.K.; Merlini, M.; Baeten, K.M.; Le Moan, N.; Petersen, M.A.; Deerinck, T.J.; Smirnoff, D.S.; Bedard, C.; Hakozaki, H.; et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat. Commun. 2012, 3, 1227. [Google Scholar] [CrossRef]
  58. Ziliotto, N.; Bernardi, F.; Piazza, F. Hemostasis components in cerebral amyloid angiopathy and Alzheimer’s disease. Neurol. Sci. 2021, 42, 3177–3188. [Google Scholar] [CrossRef]
  59. Wang, Z.; Zhang, Q.; Lin, J.R.; Jabalameli, M.R.; Mitra, J.; Nguyen, N.; Zhang, Z.D. Deep post-GWAS analysis identifies potential risk genes and risk variants for Alzheimer’s disease, providing new insights into its disease mechanisms. Sci. Rep. 2021, 11, 20511. [Google Scholar] [CrossRef]
  60. Han, M.H.; Hwang, S.I.; Roy, D.B.; Lundgren, D.H.; Price, J.V.; Ousman, S.S.; Fernald, G.H.; Gerlitz, B.; Robinson, W.H.; Baranzini, S.E.; et al. Proteomic analysis of active multiple sclerosis lesions reveals therapeutic targets. Nature 2008, 451, 1076–1081. [Google Scholar] [CrossRef]
  61. Strickland, S. Blood will out: Vascular contributions to Alzheimer’s disease. J. Clin. Investig. 2018, 128, 556–563. [Google Scholar] [CrossRef] [PubMed]
  62. Borissoff, J.I.; Spronk, H.M.; ten Cate, H. The hemostatic system as a modulator of atherosclerosis. N. Engl. J. Med. 2011, 364, 1746–1760. [Google Scholar] [CrossRef] [PubMed]
  63. Borissoff, J.I.; Spronk, H.M.H.; Heeneman, S.; ten Cate, H. Is thrombin a key player in the ‘coagulation-atherogenesis’ maze? Cardiovasc. Res. 2009, 82, 392–403. [Google Scholar] [CrossRef] [PubMed]
  64. Borissoff, J.I.; Joosen, I.A.; Versteylen, M.O.; Spronk, H.M.; ten Cate, H.; Hofstra, L. Accelerated in vivo thrombin formation independently predicts the presence and severity of CT angiographic coronary atherosclerosis. JACC Cardiovasc. Imaging 2012, 5, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  65. Colotta, F.; Sciacca, F.L.; Sironi, M.; Luini, W.; Rabiet, M.J.; Mantovani, A. Expression of monocyte chemotactic protein-1 by monocytes and endothelial cells exposed to thrombin. Am. J. Pathol. 1994, 144, 975–985. [Google Scholar]
  66. Minami, T.; Sugiyama, A.; Wu, S.Q.; Abid, R.; Kodama, T.; Aird, W.C. Thrombin and phenotypic modulation of the endothelium. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 41–53. [Google Scholar] [CrossRef]
  67. Bowen-Pope, D.F.; Raines, E.W. History of discovery: Platelet-derived growth factor. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2397–2401. [Google Scholar] [CrossRef]
  68. ten Cate, H. Tissue factor-driven thrombin generation and inflammation in atherosclerosis. Thromb. Res. 2012, 129 (Suppl. S2), S38–S40. [Google Scholar] [CrossRef]
  69. Steffel, J.; Lüscher, T.F.; Tanner, F.C. Tissue factor in cardiovascular diseases: Molecular mechanisms and clinical implications. Circulation 2006, 113, 722–731. [Google Scholar] [CrossRef]
  70. Borissoff, J.I.; Otten, J.J.; Heeneman, S.; Leenders, P.; van Oerle, R.; Soehnlein, O.; Loubele, S.T.; Hamulyak, K.; Hackeng, T.M.; Daemen, M.J.; et al. Genetic and pharmacological modifications of thrombin formation in apolipoprotein e-deficient mice determine atherosclerosis severity and atherothrombosis onset in a neutrophil-dependent manner. PLoS ONE 2013, 8, e55784. [Google Scholar] [CrossRef]
  71. Palekar, R.U.; Jallouk, A.P.; Myerson, J.W.; Pan, H.; Wickline, S.A. Inhibition of Thrombin With PPACK-Nanoparticles Restores Disrupted Endothelial Barriers and Attenuates Thrombotic Risk in Experimental Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 446–455. [Google Scholar] [CrossRef]
  72. Pingel, S.; Tiyerili, V.; Mueller, J.; Werner, N.; Nickenig, G.; Mueller, C. Thrombin inhibition by dabigatran attenuates atherosclerosis in ApoE deficient mice. Arch. Med. Sci. 2014, 10, 154–160. [Google Scholar] [CrossRef] [PubMed]
  73. Preusch, M.R.; Ieronimakis, N.; Wijelath, E.S.; Cabbage, S.; Ricks, J.; Bea, F.; Reyes, M.; van Ryn, J.; Rosenfeld, M.E. Dabigatran etexilate retards the initiation and progression of atherosclerotic lesions and inhibits the expression of oncostatin M in apolipoprotein E-deficient mice. Drug Des. Dev. Ther. 2015, 9, 5203–5211. [Google Scholar] [CrossRef] [PubMed]
  74. Wei, H.J.; Li, Y.H.; Shi, G.Y.; Liu, S.L.; Chang, P.C.; Kuo, C.H.; Wu, H.L. Thrombomodulin domains attenuate atherosclerosis by inhibiting thrombin-induced endothelial cell activation. Cardiovasc. Res. 2011, 92, 317–327. [Google Scholar] [CrossRef] [PubMed]
  75. Aoki, I.; Shimoyama, K.; Aoki, N.; Homori, M.; Yanagisawa, A.; Nakahara, K.; Kawai, Y.; Kitamura, S.I.; Ishikawa, K. Platelet-dependent thrombin generation in patients with diabetes mellitus: Effects of glycemic control on coagulability in diabetes. J. Am. Coll. Cardiol. 1996, 27, 560–566. [Google Scholar] [CrossRef] [PubMed]
  76. Chapman, J. Coagulation in inflammatory diseases of the central nervous system. Semin. Thromb. Hemost. 2013, 39, 876–880. [Google Scholar] [CrossRef]
  77. Undas, A.; Wiek, I.; Stepien, E.; Zmudka, K.; Tracz, W. Hyperglycemia is associated with enhanced thrombin formation, platelet activation, and fibrin clot resistance to lysis in patients with acute coronary syndrome. Diabetes Care 2008, 31, 1590–1595. [Google Scholar] [CrossRef]
  78. Beijers, H.J.; Ferreira, I.; Spronk, H.M.; Bravenboer, B.; Dekker, J.M.; Nijpels, G.; ten Cate, H.; Stehouwer, C.D. Body composition as determinant of thrombin generation in plasma: The Hoorn study. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2639–2647. [Google Scholar] [CrossRef] [PubMed]
  79. Lagrange, J.; Didelot, M.; Mohamadi, A.; Walton, L.A.; Bloemen, S.; de Laat, B.; Louis, H.; Thornton, S.N.; Derby, B.; Sherratt, M.J.; et al. Implication of Free Fatty Acids in Thrombin Generation and Fibrinolysis in Vascular Inflammation in Zucker Rats and Evolution with Aging. Front. Physiol. 2017, 8, 949. [Google Scholar] [CrossRef] [PubMed]
  80. Shang, J.; Chen, Z.; Wang, M.; Li, Q.; Feng, W.; Wu, Y.; Wu, W.; Graziano, M.P.; Chintala, M. Zucker Diabetic Fatty rats exhibit hypercoagulability and accelerated thrombus formation in the Arterio-Venous shunt model of thrombosis. Thromb. Res. 2014, 134, 433–439. [Google Scholar] [CrossRef]
  81. Shavit-Stein, E.; Aronovich, R.; Sylantiev, C.; Gofrit, S.G.; Chapman, J.; Dori, A. The role of thrombin in the pathogenesis of diabetic neuropathy. PLoS ONE 2019, 14, e0219453. [Google Scholar] [CrossRef]
  82. Rahadian, A.; Fukuda, D.; Salim, H.M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Shimabukuro, M.; Sata, M. Thrombin inhibition by dabigatran attenuates endothelial dysfunction in diabetic mice. Vasc. Pharmacol. 2020, 124, 106632. [Google Scholar] [CrossRef] [PubMed]
  83. Paneni, F.; Beckman, J.A.; Creager, M.A.; Cosentino, F. Diabetes and vascular disease: Pathophysiology, clinical consequences, and medical therapy: Part I. Eur. Heart J. 2013, 34, 2436–2443. [Google Scholar] [CrossRef] [PubMed]
  84. Hao, J.S.; Zhu, C.J.; Yan, B.Y.; Yan, C.Y.; Ling, R. Stimulation of KLF14/PLK1 pathway by thrombin signaling potentiates endothelial dysfunction in Type 2 diabetes mellitus. Biomed. Pharmacother. 2018, 99, 859–866. [Google Scholar] [CrossRef]
  85. Vittal Rao, H.; Bihaqi, S.W.; Iannucci, J.; Sen, A.; Grammas, P. Thrombin Signaling Contributes to High Glucose-Induced Injury of Human Brain Microvascular Endothelial Cells. J. Alzheimer’s Dis. 2021, 79, 211–224. [Google Scholar] [CrossRef]
  86. Deselms, H.; Maggio, N.; Rubovitch, V.; Chapman, J.; Schreiber, S.; Tweedie, D.; Kim, D.S.; Greig, N.H.; Pick, C.G. Novel pharmaceutical treatments for minimal traumatic brain injury and evaluation of animal models and methodologies supporting their development. J. Neurosci. Methods 2016, 272, 69–76. [Google Scholar] [CrossRef]
  87. Lindblad, C.; Thelin, E.P.; Nekludov, M.; Frostell, A.; Nelson, D.W.; Svensson, M.; Bellander, B.M. Assessment of Platelet Function in Traumatic Brain Injury-A Retrospective Observational Study in the Neuro-Critical Care Setting. Front. Neurol. 2018, 9, 15. [Google Scholar] [CrossRef]
  88. Piao, C.; Ralay Ranaivo, H.; Rusie, A.; Wadhwani, N.; Koh, S.; Wainwright, M.S. Thrombin decreases expression of the glutamate transporter GLAST and inhibits glutamate uptake in primary cortical astrocytes via the Rho kinase pathway. Exp. Neurol. 2015, 273, 288–300. [Google Scholar] [CrossRef]
  89. Li, Y.J.; Chang, G.Q.; Liu, Y.; Gong, Y.; Yang, C.; Wood, K.; Shi, F.D.; Fu, Y.; Yan, Y. Fingolimod alters inflammatory mediators and vascular permeability in intracerebral hemorrhage. Neurosci. Bull. 2015, 31, 755–762. [Google Scholar] [CrossRef]
  90. Maegele, M. Coagulopathy and Progression of Intracranial Hemorrhage in Traumatic Brain Injury: Mechanisms, Impact, and Therapeutic Considerations. Neurosurgery 2021, 89, 954–966. [Google Scholar] [CrossRef]
  91. Mortimer, J.A.; van Duijn, C.M.; Chandra, V.; Fratiglioni, L.; Graves, A.B.; Heyman, A.; Jorm, A.F.; Kokmen, E.; Kondo, K.; Rocca, W.A.; et al. Head trauma as a risk factor for Alzheimer’s disease: A collaborative re-analysis of case-control studies. EURODEM Risk Factors Research Group. Int. J. Epidemiol. 1991, 20 (Suppl. S2), S28–S35. [Google Scholar] [CrossRef] [PubMed]
  92. Nemetz, P.N.; Leibson, C.; Naessens, J.M.; Beard, M.; Kokmen, E.; Annegers, J.F.; Kurland, L.T. Traumatic brain injury and time to onset of Alzheimer’s disease: A population-based study. Am. J. Epidemiol. 1999, 149, 32–40. [Google Scholar] [CrossRef] [PubMed]
  93. 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]
  94. 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]
  95. Luo, J.; Wu, X.; Liu, H.; Cui, W.; Guo, W.; Guo, K.; Guo, H.; Tao, K.; Li, F.; Shi, Y.; et al. Antagonism of Protease-Activated Receptor 4 Protects against Traumatic Brain Injury by Suppressing Neuroinflammation via Inhibition of Tab2/NF-κB Signaling. Neurosci. Bull. 2021, 37, 242–254. [Google Scholar] [CrossRef]
  96. Shlosberg, D.; Benifla, M.; Kaufer, D.; Friedman, A. Blood-brain barrier breakdown as a therapeutic target in traumatic brain injury. Nat. Rev. Neurol. 2010, 6, 393–403. [Google Scholar] [CrossRef]
  97. Alabanza, L.M.; Bynoe, M.S. Thrombin induces an inflammatory phenotype in a human brain endothelial cell line. J. Neuroimmunol. 2012, 245, 48–55. [Google Scholar] [CrossRef]
  98. 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]
  99. Arai, T.; Guo, J.P.; McGeer, P.L. Proteolysis of non-phosphorylated and phosphorylated tau by thrombin. J. Biol. Chem. 2005, 280, 5145–5153. [Google Scholar] [CrossRef]
  100. 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]
  101. Zamolodchikov, D.; Chen, Z.L.; Conti, B.A.; Renne, T.; Strickland, S. Activation of the factor XII-driven contact system in Alzheimer’s disease patient and mouse model plasma. Proc. Natl. Acad. Sci. USA 2015, 112, 4068–4073. [Google Scholar] [CrossRef] [PubMed]
  102. Mhatre, M.; Hensley, K.; Nguyen, A.; Grammas, P. Chronic thrombin exposure results in an increase in apolipoprotein-E levels. J. Neurosci. Res. 2006, 84, 444–449. [Google Scholar] [CrossRef] [PubMed]
  103. Mhatre, M.; Nguyen, A.; Kashani, S.; Pham, T.; Adesina, A.; Grammas, P. Thrombin, a mediator of neurotoxicity and memory impairment. Neurobiol. Aging 2004, 25, 783–793. [Google Scholar] [CrossRef]
  104. Han, C.; Xia, X.; Jiao, S.; Li, G.; Ran, Q.; Yao, S. Tripartite motif containing protein 37 involves in thrombin stimulated BV-2 microglial cell apoptosis and interleukin 1beta release. Biochem. Biophys. Res. Commun. 2019, 516, 1252–1257. [Google Scholar] [CrossRef] [PubMed]
  105. Ye, X.; Zuo, D.; Yu, L.; Zhang, L.; Tang, J.; Cui, C.; Bao, L.; Zan, K.; Zhang, Z.; Yang, X.; et al. ROS/TXNIP pathway contributes to thrombin induced NLRP3 inflammasome activation and cell apoptosis in microglia. Biochem. Biophys. Res. Commun. 2017, 485, 499–505. [Google Scholar] [CrossRef] [PubMed]
  106. Huang, C.; Ma, R.; Sun, S.; Wei, G.; Fang, Y.; Liu, R.; Li, G. JAK2-STAT3 signaling pathway mediates thrombin-induced proinflammatory actions of microglia in vitro. J. Neuroimmunol. 2008, 204, 118–125. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, D.Y.; Park, K.W.; Jin, B.K. Thrombin induces neurodegeneration and microglial activation in the cortex in vivo and in vitro: Proteolytic and non-proteolytic actions. Biochem. Biophys. Res. Commun. 2006, 346, 727–738. [Google Scholar] [CrossRef]
  108. Yang, Y.; Zhang, M.; Kang, X.; Jiang, C.; Zhang, H.; Wang, P.; Li, J. Thrombin-induced microglial activation impairs hippocampal neurogenesis and spatial memory ability in mice. Behav. Brain Funct. 2015, 11, 30. [Google Scholar] [CrossRef]
  109. 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]
  110. Yin, M.; Chen, Z.; Ouyang, Y.; Zhang, H.; Wan, Z.; Wang, H.; Wu, W.; Yin, X. Thrombin-induced, TNFR-dependent miR-181c downregulation promotes MLL1 and NF-κB target gene expression in human microglia. J. Neuroinflamm. 2017, 14, 132. [Google Scholar] [CrossRef]
  111. Lin, C.C.; Lee, I.T.; Wu, W.B.; Liu, C.J.; Hsieh, H.L.; Hsiao, L.D.; Yang, C.C.; Yang, C.M. Thrombin mediates migration of rat brain astrocytes via PLC, Ca2+, CaMKII, PKCalpha, and AP-1-dependent matrix metalloproteinase-9 expression. Mol. Neurobiol. 2013, 48, 616–630. [Google Scholar] [CrossRef] [PubMed]
  112. Cavanaugh, K.P.; Gurwitz, D.; Cunningham, D.D.; Bradshaw, R.A. Reciprocal modulation of astrocyte stellation by thrombin and protease nexin-1. J. Neurochem. 1990, 54, 1735–1743. [Google Scholar] [CrossRef] [PubMed]
  113. Birukova, A.A.; Birukov, K.G.; Smurova, K.; Adyshev, D.; Kaibuchi, K.; Alieva, I.; Garcia, J.G.; Verin, A.D. Novel role of microtubules in thrombin-induced endothelial barrier dysfunction. FASEB J. 2004, 18, 1879–1890. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, D.Z.; Ander, B.P.; Xu, H.; Shen, Y.; Kaur, P.; Deng, W.; Sharp, F.R. Blood-brain barrier breakdown and repair by Src after thrombin-induced injury. Ann. Neurol. 2010, 67, 526–533. [Google Scholar] [CrossRef] [PubMed]
  115. 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, 248–255. [Google Scholar] [CrossRef]
  116. Mongkhon, P.; Naser, A.Y.; Fanning, L.; Tse, G.; Lau, W.C.Y.; Wong, I.C.K.; Kongkaew, C. Oral anticoagulants and risk of dementia: A systematic review and meta-analysis of observational studies and randomized controlled trials. Neurosci. Biobehav. Rev. 2019, 96, 1–9. [Google Scholar] [CrossRef]
  117. Ding, M.; Fratiglioni, L.; Johnell, K.; Santoni, G.; Fastbom, J.; Ljungman, P.; Marengoni, A.; Qiu, C. Atrial fibrillation, antithrombotic treatment, and cognitive aging: A population-based study. Neurology 2018, 91, e1732–e1740. [Google Scholar] [CrossRef]
  118. Field, T.S.; Weijs, B.; Curcio, A.; Giustozzi, M.; Sudikas, S.; Katholing, A.; Wallenhorst, C.; Weitz, J.I.; Cohen, A.T.; Martinez, C. Incident Atrial Fibrillation, Dementia and the Role of Anticoagulation: A Population-Based Cohort Study. Thromb. Haemost. 2019, 119, 981–991. [Google Scholar] [CrossRef]
  119. Silva, R.; Miranda, C.M.; Liu, T.; Tse, G.; Roever, L. Atrial Fibrillation and Risk of Dementia: Epidemiology, Mechanisms, and Effect of Anticoagulation. Front. Neurosci. 2019, 13, 18. [Google Scholar] [CrossRef]
  120. Bergamaschini, L.; Rossi, E.; Storini, C.; Pizzimenti, S.; Distaso, M.; Perego, C.; De Luigi, A.; Vergani, C.; De Simoni, M.G. Peripheral treatment with enoxaparin, a low molecular weight heparin, reduces plaques and beta-amyloid accumulation in a mouse model of Alzheimer’s disease. J. Neurosci. 2004, 24, 4181–4186. [Google Scholar] [CrossRef]
  121. Cortes-Canteli, M.; Kruyer, A.; Fernandez-Nueda, I.; Marcos-Diaz, A.; Ceron, C.; Richards, A.T.; Jno-Charles, O.C.; Rodriguez, I.; Callejas, S.; Norris, E.H.; et al. Long-Term Dabigatran Treatment Delays Alzheimer’s Disease Pathogenesis in the TgCRND8 Mouse Model. J. Am. Coll. Cardiol. 2019, 74, 1910–1923. [Google Scholar] [CrossRef] [PubMed]
  122. 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] [CrossRef]
  123. Friberg, L.; Rosenqvist, M. Less dementia with oral anticoagulation in atrial fibrillation. Eur. Heart J. 2018, 39, 453–460. [Google Scholar] [CrossRef]
  124. Mongkhon, P.; Fanning, L.; Lau, W.C.Y.; Tse, G.; Lau, K.K.; Wei, L.; Kongkaew, C.; Wong, I.C.K. Oral anticoagulant and reduced risk of dementia in patients with atrial fibrillation: A population-based cohort study. Heart Rhythm 2020, 17, 706–713. [Google Scholar] [CrossRef]
  125. Jacobs, V.; May, H.T.; Bair, T.L.; Crandall, B.G.; Cutler, M.J.; Day, J.D.; Mallender, C.; Osborn, J.S.; Stevens, S.M.; Weiss, J.P.; et al. Long-Term Population-Based Cerebral Ischemic Event and Cognitive Outcomes of Direct Oral Anticoagulants Compared with Warfarin among Long-term Anticoagulated Patients for Atrial Fibrillation. Am. J. Cardiol. 2016, 118, 210–214. [Google Scholar] [CrossRef]
  126. Cadogan, S.L.; Powell, E.; Wing, K.; Wong, A.Y.; Smeeth, L.; Warren-Gash, C. Anticoagulant prescribing for atrial fibrillation and risk of incident dementia. Heart 2021, 107, 1898–1904. [Google Scholar] [CrossRef] [PubMed]
  127. Cheng, W.; Liu, W.; Li, B.; Li, D. Relationship of Anticoagulant Therapy with Cognitive Impairment among Patients with Atrial Fibrillation: A Meta-Analysis and Systematic Review. J. Cardiovasc. Pharmacol. 2018, 71, 380–387. [Google Scholar] [CrossRef]
  128. Zeng, D.; Jiang, C.; Su, C.; Tan, Y.; Wu, J. Anticoagulation in atrial fibrillation and cognitive decline: A systematic review and meta-analysis. Medicine 2019, 98, e14499. [Google Scholar] [CrossRef] [PubMed]
  129. Ho, B.L.; Hsieh, S.W.; Chou, P.S.; Yang, Y.H. Effects of Dabigatran on Dementia Pathogenesis and Neuropsychological Function: A Review. J. Alzheimer’s Dis. 2022, 86, 1589–1601. [Google Scholar] [CrossRef]
  130. Grossmann, K. Direct Oral Anticoagulants (DOACs) for Therapeutic Targeting of Thrombin, a Key Mediator of Cerebrovascular and Neuronal Dysfunction in Alzheimer’s Disease. Biomedicines 2022, 10, 1890. [Google Scholar] [CrossRef]
  131. Ruff, C.T.; Giugliano, R.P.; Braunwald, E.; Hoffman, E.B.; Deenadayalu, N.; Ezekowitz, M.D.; Camm, A.J.; Weitz, J.I.; Lewis, B.S.; Parkhomenko, A.; et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: A meta-analysis of randomised trials. Lancet 2014, 383, 955–962. [Google Scholar] [CrossRef] [PubMed]
  132. Graham, D.J.; Reichman, M.E.; Wernecke, M.; Zhang, R.; Southworth, M.R.; Levenson, M.; Sheu, T.C.; Mott, K.; Goulding, M.R.; Houstoun, M.; et al. Cardiovascular, bleeding, and mortality risks in elderly Medicare patients treated with dabigatran or warfarin for nonvalvular atrial fibrillation. Circulation 2015, 131, 157–164. [Google Scholar] [CrossRef] [PubMed]
  133. Graham, D.J.; Reichman, M.E.; Wernecke, M.; Hsueh, Y.H.; Izem, R.; Southworth, M.R.; Wei, Y.; Liao, J.; Goulding, M.R.; Mott, K.; et al. Stroke, Bleeding, and Mortality Risks in Elderly Medicare Beneficiaries Treated with Dabigatran or Rivaroxaban for Nonvalvular Atrial Fibrillation. JAMA Intern. Med. 2016, 176, 1662–1671. [Google Scholar] [CrossRef] [PubMed]
  134. Ferro, C.J.; Solkhon, F.; Jalal, Z.; Al-Hamid, A.M.; Jones, A.M. Relevance of physicochemical properties and functional pharmacology data to predict the clinical safety profile of direct oral anticoagulants. Pharmacol. Res. Perspect. 2020, 8, e00603. [Google Scholar] [CrossRef]
  135. Marinescu, M.; Sun, L.; Fatar, M.; Neubauer, A.; Schad, L.; van Ryn, J.; Lehmann, L.; Veltkamp, R. Cerebral Microbleeds in Murine Amyloid Angiopathy: Natural Course and Anticoagulant Effects. Stroke 2017, 48, 2248–2254. [Google Scholar] [CrossRef]
  136. Michael, N.; Grigoryan, M.M.; Kilday, K.; Sumbria, R.K.; Vasilevko, V.; van Ryn, J.; Cribbs, D.H.; Paganini-Hill, A.; Fisher, M.J. Effects of Dabigatran in Mouse Models of Aging and Cerebral Amyloid Angiopathy. Front. Neurol. 2019, 10, 966. [Google Scholar] [CrossRef]
  137. Cummings, J.L.; Morstorf, T.; Zhong, K. Alzheimer’s disease drug-development pipeline: Few candidates, frequent failures. Alzheimer’s Res. Ther. 2014, 6, 37. [Google Scholar] [CrossRef]
  138. Tolar, M.; Abushakra, S.; Sabbagh, M. The path forward in Alzheimer’s disease therapeutics: Reevaluating the amyloid cascade hypothesis. Alzheimer’s Dement. 2020, 16, 1553–1560. [Google Scholar] [CrossRef]
  139. Servick, K. Alzheimer’s drug approved despite murky results. Science 2021, 372, 1141. [Google Scholar] [CrossRef]
  140. van Dyck, C.H.; Swanson, C.J.; Aisen, P.; Bateman, R.J.; Chen, C.; Gee, M.; Kanekiyo, M.; Li, D.; Reyderman, L.; Cohen, S.; et al. Lecanemab in Early Alzheimer’s Disease. N. Engl. J. Med. 2023, 388, 9–21. [Google Scholar] [CrossRef]
  141. Hampel, H.; Caraci, F.; Cuello, A.C.; Caruso, G.; Nisticò, R.; Corbo, M.; Baldacci, F.; Toschi, N.; Garaci, F.; Chiesa, P.A.; et al. A Path Toward Precision Medicine for Neuroinflammatory Mechanisms in Alzheimer’s Disease. Front. Immunol. 2020, 11, 456. [Google Scholar] [CrossRef] [PubMed]
  142. Lim, G.P.; Yang, F.; Chu, T.; Chen, P.; Beech, W.; Teter, B.; Tran, T.; Ubeda, O.; Ashe, K.H.; Frautschy, S.A.; et al. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J. Neurosci. 2000, 20, 5709–5714. [Google Scholar] [CrossRef] [PubMed]
  143. McGeer, P.L.; Schulzer, M.; McGeer, E.G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease: A review of 17 epidemiologic studies. Neurology 1996, 47, 425–432. [Google Scholar] [CrossRef]
  144. Stewart, W.F.; Kawas, C.; Corrada, M.; Metter, E.J. Risk of Alzheimer’s disease and duration of NSAID use. Neurology 1997, 48, 626–632. [Google Scholar] [CrossRef] [PubMed]
  145. Hayden, K.M.; Zandi, P.P.; Khachaturian, A.S.; Szekely, C.A.; Fotuhi, M.; Norton, M.C.; Tschanz, J.T.; Pieper, C.F.; Corcoran, C.; Lyketsos, C.G.; et al. Does NSAID use modify cognitive trajectories in the elderly? The Cache County study. Neurology 2007, 69, 275–282. [Google Scholar] [CrossRef] [PubMed]
  146. Vlad, S.C.; Miller, D.R.; Kowall, N.W.; Felson, D.T. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology 2008, 70, 1672–1677. [Google Scholar] [CrossRef]
  147. Zhang, C.; Wang, Y.; Wang, D.; Zhang, J.; Zhang, F. NSAID Exposure and Risk of Alzheimer’s Disease: An Updated Meta-Analysis from Cohort Studies. Front. Aging Neurosci. 2018, 10, 83. [Google Scholar] [CrossRef]
  148. Cummings, J.L.; Tong, G.; Ballard, C. Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. J. Alzheimer’s Dis. 2019, 67, 779–794. [Google Scholar] [CrossRef]
Figure 1. Thrombin-mediated neuroinflammation in neurodegenerative disease. Thrombin, produced by brain endothelial cells, acts as a regulator of neuroinflammatory processes in the brain, including inflammatory cytokines, matrix metalloproteinases (MMPs), reactive oxygen species (ROS), and nitric oxide (NO). Together, these inflammatory mediators can damage a healthy neuron and drive the neurodegeneration seen in Alzheimer’s disease (AD) and other neurodegenerative disorders.
Figure 1. Thrombin-mediated neuroinflammation in neurodegenerative disease. Thrombin, produced by brain endothelial cells, acts as a regulator of neuroinflammatory processes in the brain, including inflammatory cytokines, matrix metalloproteinases (MMPs), reactive oxygen species (ROS), and nitric oxide (NO). Together, these inflammatory mediators can damage a healthy neuron and drive the neurodegeneration seen in Alzheimer’s disease (AD) and other neurodegenerative disorders.
Cells 12 01222 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iannucci, J.; Grammas, P. Thrombin, a Key Driver of Pathological Inflammation in the Brain. Cells 2023, 12, 1222. https://doi.org/10.3390/cells12091222

AMA Style

Iannucci J, Grammas P. Thrombin, a Key Driver of Pathological Inflammation in the Brain. Cells. 2023; 12(9):1222. https://doi.org/10.3390/cells12091222

Chicago/Turabian Style

Iannucci, Jaclyn, and Paula Grammas. 2023. "Thrombin, a Key Driver of Pathological Inflammation in the Brain" Cells 12, no. 9: 1222. https://doi.org/10.3390/cells12091222

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