Next Article in Journal
Plasma Biology
Next Article in Special Issue
Valproic Acid Reduces Vasospasm through Modulation of Akt Phosphorylation and Attenuates Neuronal Apoptosis in Subarachnoid Hemorrhage Rats
Previous Article in Journal
Cell Proliferation in the Piriform Cortex of Rats with Motor Cortex Ablation Treated with Growth Hormone and Rehabilitation
Previous Article in Special Issue
The Pathogenesis of Hydrocephalus Following Aneurysmal Subarachnoid Hemorrhage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH)

1
Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
2
Department of Neurosurgery, Helios-Amper Klinikum Dachau, Krankenhausstr. 15, 85221 Dachau, Germany
3
Department of Neurosurgery, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
*
Authors to whom correspondence should be addressed.
Joint first authors with equal contributions.
Int. J. Mol. Sci. 2021, 22(11), 5442; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115442
Submission received: 17 March 2021 / Revised: 30 April 2021 / Accepted: 18 May 2021 / Published: 21 May 2021

Abstract

:
Aneurysmal subarachnoid hemorrhage (aSAH) remains a disease with high mortality and morbidity. Since treating vasospasm has not inevitably led to an improvement in outcome, the actual emphasis is on finding neuroprotective therapies in the early phase following aSAH to prevent secondary brain injury in the later phase of disease. Within the early phase, neuroinflammation, thromboinflammation, disturbances in brain metabolism and early neuroprotective therapies directed against delayed cerebral ischemia (DCI) came into focus. Herein, the role of neuroinflammation, thromboinflammation and metabolism in aSAH is depicted. Potential neuroprotective strategies regarding neuroinflammation target microglia activation, metalloproteases, autophagy and the pathway via Toll-like receptor 4 (TLR4), high mobility group box 1 (HMGB1), NF-κB and finally the release of cytokines like TNFα or IL-1. Following the link to thromboinflammation, potential neuroprotective therapies try to target microthrombus formation, platelets and platelet receptors as well as clot clearance and immune cell infiltration. Potential neuroprotective strategies regarding metabolism try to re-balance the mismatch of energy need and supply following aSAH, for example, in restoring fuel to the TCA cycle or bypassing distinct energy pathways. Overall, this review addresses current neuroprotective strategies in aSAH, hopefully leading to future translational therapy options to prevent secondary brain injury.

1. Introduction

Aneurysmal subarachnoid hemorrhage (aSAH) is a complex cerebrovascular disease with profound systemic complications, accounting for about 5% of all strokes. The worldwide incidence of aSAH is approximately 700,000 person–years with an overall mortality of about 40% [1,2]. Brain injury following aSAH is multimodal and occurs directly, as early brain injury, however, also secondarily, as delayed brain injury [3]. Angiographic cerebral vasospasm (CVS) occurs in approximately 70% of patients during the first 2 weeks after aSAH, but the incidence of delayed cerebral ischemia (DCI) is only around 30%, with DCI remaining the major cause of morbidity and mortality among patients who survive the initial treatment of the ruptured aneurysm [4,5]. Several mechanisms during the acute phase of SAH contribute to DCI and poor outcome. These include neuroinflammation, microthrombosis, cortical spreading depolarizations, disrupted integrity of the blood–brain barrier (BBB), microvascular dysfunction and metabolic derangement [6,7,8,9].
Despite the extensive advances in experimental research, especially in recent years, translational clinical therapy and data are still lacking, persistently motivating researchers all over the world to find new strategies for neuroprotective therapy following aSAH. The main goal of this review was to summarize the role of neuroinflammation, thromboinflammation and metabolism following aSAH with a special focus on new neuroprotective targets in these fields, possibly leading to translational new approaches in clinical neuroprotective therapy.

2. Role of Neuroinflammation in aSAH

2.1. Activation of the Immune System

The initial global hypoperfusion after aSAH leads to inflammatory processes, which occur in blood vessels as well as in cerebrospinal fluid (CSF). While talking about inflammation in a non-infected environment, persistent actions of the innate immune system were predominantly found [10,11,12].
Peripheral monocytes invade the brain as macrophages. Lymphocytes, as part of the innate and adapted immunity, are highly activated in contrast to B- and T-cells as products of the adapted immune system, which are just rarely upregulated [13]. Since macrophages and neutrophils enter the subarachnoid space, they degranulate, whereby inflammatory factors are released [14].
An early gain of neutrophils, already 3 days after aSAH, was found to be associated with a later vasospasm. Therefore, it can be concluded, that the mechanism of neutrophil signaling ameliorates vasospasm.
Inflammatory cells and associated cytokines as well as their receptors are upregulated in the subarachnoid space, while entering from within the blood vessels and acting on vascular walls formed by the increased reactivity of microvessels to post-hemorrhagic CSF [15].
An increased permeability of the BBB enables cytokines to reach the brain parenchyma and also circulating immune cells to access the perivascular spaces and reach the brain interstitium [16,17]. Additionally, an increased cytokine production as a response to injured brain parenchyma and the increased permeability of the BBB leads to global cerebral edema.

2.2. Acute Events Following aSAH

After the initial aneurysm rupture, blood leaks into the subarachnoid space. The breakdown of red blood cells and degradation over time leads to the deposition of hemoglobin. As a result of red blood cell breakdown, methemoglobin, heme and hemin can lead to activation of Toll-like receptor 4 (TLR4), which signals inflammatory cascades that damage neurons and white matter [18]. Due to the neuroimmunological damage of neurons, a possible link with metabolic derangement may exist. Via the release of redox-active iron, Hemin has been linked to alter the balance of oxidants and anti-oxidants. The redox-active ion produces superoxide and hydroxyl radicals as well as lipid peroxidation while depleting anti-oxidant stores such as nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione [18,19].
In addition, immunomodulatory cells, notably microglia, are activated due to leaking blood after aneurysm rupture. These cells trigger the upregulation of numerous cell adhesion molecules within endothelial cells, which subsequently allows a multitude of inflammatory cells to bind and enter the subarachnoid space [18,20]. Once these inflammatory cells, such as macrophages and neutrophils, enter the subarachnoid space, they phagocytize the extravasated, degrading blood cells in an effort to clear free hemoglobin and therefore promote neurostability and recovery. By the binding of hemoglobin to haptoglobin, a rapid engulfment by immune cells is facilitated [18].

2.3. Subacute/Chronic Events Following aSAH

As described above, peripheral immune cells such as neutrophils and macrophages are attracted to clear free hemoglobin after aneurysm rupture. Moreover, they might also become trapped in the subarachnoid space due to alterations in CSF flow and the restoration of the endothelial tight junction barrier. During degranulation in the subarachnoid space, macrophages and neutrophils release a multitude of inflammatory factors, including endothelins and oxidative radicals. Further downstream, these factors can cause inflammation-induced vasoconstriction, meningitis and cerebritis [21]. It remains unclear whether neutrophils and macrophages are passing through an intact or disrupted BBB while being recruited.

2.4. Microglia

The microglia can be understood as the on-site phagocytes of the central nervous system (CNS), which are able to provoke an upregulation of inflammatory cytokines, especially of interleukins IL-1ß and IL-6 as well as of tumor necrosis factor alpha (TNFα) as a response to infection or even hemorrhage due to an accumulation near the vessel rupture with release into both serum and CSF [22,23,24].
This increased production causes the destabilization of the blood–brain barrier by a wave of intraparenchymal inflammation, which causes neuronal injury where it has typical pro-inflammatory cytokines, even if aSAH is primary a bleeding outside the brain parenchyma. The breakdown of red blood cell products and its release of inflammatory cytokeratins could be understood as a trigger of vasospasm and tissue injury [14].
During the early phase after aSAH, as already mentioned above, the breakdown and degradation of red blood cells leads to a hemoglobin detachment. Subsequently, methemoglobin, heme and hemin facilitate a neuroinflammatory reaction as they are able to activate TLR4, which are expressed in microglia [25,26]. TLR4 generates among others an inflammatory response by interacting with the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), which results in damaging nerve tissue [27]. In summary, different pathways linked to TLR4 activation were found. Therefore, new strategies for immunotherapy that target microglia and TLR4 signaling should be investigated [28].
Microglia accumulation plays a significant role in the cerebral spreading of inflammation [29] as it can be a significant target for treatment strategies. The amount of neuronal cell death can be reduced by microglia depletion. The intracerebral accumulation of inflammatory cells occurs between day 4 and 28 after aSAH, contributing to delayed brain injury [30].

2.5. Endocannabinoids

Endocannabinoids (eCBs) are lipid-soluble molecules and the endogenous analogs of the psychoactive constituents of cannabis plants. They are produced on demand in response to increased intracellular calcium levels. Arachidonoylethanolamine, also known as anandamide (AEA), and 2-arachidonoyl-glycerol (2-AG) are the main representatives of eCBs. The endocannabinoid system (ECS) is described as a complex lipid network consisting of cannabinoid receptors (CBRs), endogenous ligands and the enzymes involved in endocannabinoid degradation and synthesis [31,32]. The ECS has been implicated as an important regulatory component by the regulation of the local CBF and by the maintenance of the BBB and related transport proteins [32].
All major cell types involved in cerebrovascular control pathways (i.e., smooth muscle, endothelium, neurons, astrocytes, pericytes, microglia and leukocytes) are capable of synthesizing eCBs and/or the expression of their target proteins like the cannabinoid type 1 receptor (CB1R) and cannabinoid type 2 receptor (CB2R).
This leads to the hypothesis, that the ECS may importantly modulate the regulation of cerebral circulation in physiological and pathophysiological conditions. Experimental data suggest that the direct effect of cannabinoids on cerebral vessels is vasodilatation, at least partially mediated by CB1R. It was shown that, for certain cerebrovascular pathologies like SAH and traumatic as well as ischemic brain injury, the activation of CB2R (and to date unidentified non-CB1R/non-CB2R) receptors may improve brain blood perfusion by attenuating vascular inflammation [32].
Microglia in the homeostatic “resting” state synthesize 2-AG and AEA and express the cannabinoid receptors CB1R and CB2R at low levels. When activated, microglia significantly increase their synthesis of eCBs and upregulate their expression of CB2R, making them produce more neuroprotective and less pro-inflammatory factors [33].
Both synthetic cannabinoids and eCBs were shown to inhibit TNFα release and the release of other cytokines like IL-1α, IL-1β and IL-6 [32].

2.6. Metalloproteinases

Metalloproteinases (MMPs) are a family of proteases consisting of multiple subtypes. MMP-9 is the most widely investigated metalloproteinase and has been shown to be responsible for the degradation of tight junction proteins, which are substantial in the maintenance of BBB integrity. Clinical studies of aSAH have reported an elevation of MMP-9 levels in brain tissue, serum and CSF as well as in the vessel wall [34]. Therefore, MMP-9 might be a possible target for neuroprotective therapies by restoring BBB integrity or even prevent BBB disruption in the first place.

2.7. High Mobility Group Box 1

The high mobility group box 1 (HMGB1) protein is a pro-inflammatory-like cytokine with an initiator role in neuroinflammation, that has been implicated in traumatic brain injury (TBI) as well as in early brain injury (EBI) after aSAH. An extracellular overexpression of HMGB1 following aSAH has been described. Once reaching the extracellular milieu, HMGB1 serves as a damage-associated molecular pattern (DAMP) protein and exerts an inflammatory response, engaging several inflammatory mediators. After being released from neurons and astrocytes, HMGB1 begins the production of several inflammatory markers, including TNFα, IL-6 and IL-1β [35]. Extracellular HMGB1 interacts with TLR4 and the receptor for advanced glycation end products (RAGE) to initiate cell migration and the production of cytokines [35,36].
HMB1 acts as an inflammatory mediator due to the activation of neuroinflammatory cascades and rupture of the BBB [37,38]. The increased HMGB1 leads to an aggravated inflammation and upregulation of inflammatory mediators via TLRs/NF-kB signaling cascades [39]. An early increase following aSAH with its maximum peak after one day and its proinflammatory role was demonstrated in experimental aSAH by the verification of increased TLR4, IL-1ß as well as NF-kB after the administration of recombinant HMGB1 [40]. By activating Janus-Kinase (JAK2)/signal transducer and activator of transcription (STAT3), it contributes to EBI. Therefore, the JAK2/STAT3 inhibitor AG490 decreased the HMGB1 expression as well as its nuclear to cytosol translocation, which leads to ameliorated EBI in experimental aSAH [41].
The experimental administration of a HGMB1 antibody demonstrated an attenuated progression of CVS due to a reduced upregulation of inflammatory molecules such as TNFα, TLR4 and IL-6 [38]. Accordingly, glycyrrhizin (a HGMB1 inhibiting molecule) reduced CVS by the downregulation of proinflammatory cytokines (IL1-ß, IL-6 as well as TNFα) [42].
Even though no clinical data have been provided to date, targeting the HMGB1 pathways may be a promising therapeutic approach [39]. Elevated serum HMGB1 levels in aSAH-patients from day 1 remained elevated until day 13 in patients developing CVS, reflecting a biomarker potential of HMGB1 [43].

2.8. Autophagy and NF-κB-Mediated Inflammation

SAH following aneurysm rupture is related to an affected function of the autophagy lysosomal system. Autophagy implies “self-eating” and can be understood as a process of lysosomal degradation of no longer usable cellular components. Therefore, it sets an important element of cellular metabolism [44].
Among the most serious consequences of intracranial aneurysm rupture are EBI, delayed brain injury and CVS, all associated with an impaired function of the autophagy-lysosomal system [44]. Aneurysm walls are usually characterized by an active inflammatory response, and NF-κB has been identified as the main transcription factor regulating the induction of inflammation-related genes in intracranial aneurysm lesions [45]. Moreover, NF-κB, which is a pivotal factor controlling inflammation, is regulated by autophagy-related proteins, and autophagy is regulated by NF-κB signaling. NF-κB denotes a family of transcription factors playing an important role in many cellular activities including proliferation, response to stress, immune response, apoptosis and inflammation [46]. Under stress conditions, NF-κB translocates to the nucleus, where it stimulates or represses the transcription of many genes. The activation of NF-κB in basal (normal) conditions can be promoted by two separate signaling pathways, the canonical and non-canonical pathway. The basal pathway can be stimulated by various factors, but in general the canonical pathway is activated by TNFα and IL-1β, as well as other cytokines via binding to their specific receptors [47]. The non-canonical pathway gets activated by a limited set of ligands, including those belonging to the TNF family and TNF-related factors. NF-κB, in an elevated activated form, generates the expression of several pro-inflammatory proteins, including cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE-2) and molecules that facilitate the recruitment and adhesion of macrophages [45,48]. Once they have entered the vessel wall through new vasa vasorum, macrophages release other pro-inflammatory molecules, including TNFα, IL-1β, MMPs as well as other proteases, which finally results in the remodeling of the structure of the vessel wall in combination with other molecules [45,47,48]. An antiapoptotic effect was demonstrated due to the activation of autophagy with the immunosuppressive agent rapamycin (also named sirolimus), which is known as an inducer of autophagy undergoing several clinical trials in vascular diseases. In experimental aSAH, the activation of autophagy leads to decreased degree of CVS [49]. In contrast, an autophagy inhibition resulted in the deterioration of neurological deficits [50]. Therefore, autophagy and the NF-κB-mediated pathway might be other potential targets for neuroprotective therapies following aSAH, with special regard to the balance between physiologic and pathologic mechanisms of autophagy.

2.9. Meningeal Lymphatics

Caused by aSAH, blood, and accordingly red blood cell degradation products, rapidly enter the subarachnoid space and therefore the paravascular pathway, resulting in distinctly perivascular neuroinflammation [51]. A brain-wide pathway connecting perivascular spaces with CSF is the recently rediscovered system of lymphatic vessels, known as the glymphatic system, which is the utmost important in the clearance of metabolic waste products [52]. In addition to clots, lysis and phagocytosis by macrophages and neutrophils, meningeal lymphatic vessels drain CSF macromolecules and immune cells to cervical lymph nodes [53,54].
As mentioned earlier, after aSAH microglia is activated by the upregulation of inflammatory cytokines including TNFα, IL-1ß and IL-6, these are aggravated by the ablation of meningeal lymphatics, which leads to exacerbated neuroinflammation and neurological deficits. Therefore, the lymphatic system provides another promising opportunity in the development of therapeutic strategies regarding the treatment of brain injury after aSAH [55].

2.10. Novel Therapies Targeting Neuroinflammation

Inflammatory events that happen in the subarachnoid space can be divided into (1) cellular inflammation and (2) molecular inflammation. In addition, inflammatory processes take place in the three compartments (1) subarachnoid space, (2) vascular lumen and (3) cerebral parenchyma [3]. On the one hand, cellular components leave the blood vessels and enter the subarachnoid space. On the other hand, cellular and molecular factors act on the vascular walls. These findings raise the question of whether inflammation in the CSF is an outside–in or an inside–out reaction [3].
Considering an inflammatory response due to aSAH, a neuroprotective effect of early treatment with anti-inflammatory drugs as corticosteroids showed a diminished brain damage and a reduced mortality in a rat model [56]. Even a non-significant trend to improved neurological recovery could be demonstrated. However, several clinical studies could not confirm a beneficial effect of treatment with corticosteroids after aSAH in regard to CVS [57,58].
Another promising agent is human albumin, as it is known for anti-inflammatory properties [59,60]. The ALISAH study demonstrated neuroprotective effects due to a lower incidence of CVS and a trend of better neurological outcome [61].
A neuroprotective effect and reduced inflammation were recently demonstrated by the application of an IL-1 receptor antagonist (IL-1Ra) [62,63]. IL-6 was reduced in plasma and CSF, as IL-1Ra was able to cross the BBB. Therefore, IL-1Ra could be a safe neuroprotective agent.
Regarding useful potential biomarkers in the treatment of SAH patients as well as for prediction of outcome and post-SAH infections, serum IL-10 might be an additional useful parameter. SAH patients, who developed any kind of infection, CVS or chronic hydrocephalus, had significantly higher serum IL-10 levels compared to controls. In addition, discharged SAH patients with poor clinical outcome (modified ranking scale 3–6 or Glasgow outcome scale 1–3) revealed significantly higher serum IL-10 levels [64]. Another promising predictive biomarker correlating with clinical neurological outcome might be chemokine C-C motif ligand 5 (CCL5). Systemic and CSF CCL5 levels in aSAH patients on day 1 and day 7 could be independently associated with the clinical outcome at discharge. Therefore, CCL5 might also be another target for neuroprotective strategies in aSAH [65].
A potential neuroprotective agent regarding endocannabinoids might be the selective CB2R agonist JWH133. The application of JWH133 after experimental SAH achieved a protection of the BBB, shown in reduced brain edema and the attenuation of neurological outcome by suppressed leukocyte infiltration through TGF-1β upregulation and reduced neuronal apoptosis [66,67]. The same agent (JWH133) was also able to attenuate acute neurogenic pulmonary edema by preventing neutrophil migration after experimental SAH in rats [68]. In addition, the production of TNFα has been suppressed by cannabinoids leading to reduced CVS and brain ischemia after SAH [32]. In conflict with these results, a retrospective analysis showed a poor clinical outcome in SAH patients with verified cannabis consumption by the increased onset of delayed cerebral ischemia [69].
The challenge with regard to modulating inflammation is the fact that inflammation is often observed to be biphasic in nature, with elements that are both protective as well as deleterious. Identifying this temporal relationship and when to target involved pathways for therapeutic benefit remains a substantial challenge. Advanced neuroimaging may offer a viable option to detect biphasic peaks in the neuroinflammatory cascade. Finally, utilizing our current knowledge of SAH pathophysiology offers clear advantages therapeutically.
Table 1 summarizes new potential neuroprotective therapies in aSAH targeting neuroinflammation performed over the last 5 years. Despite the immense increase in the number of experimental, translational clinical data, some are still missing. A complementary collection of clinical neuroprotective therapies targeting neuroinflammation was nicely summarized by de Oliveira and Macdonald in 2018 [70]. Studies on neuroprotective therapies targeting neuroinflammation in aSAH older than 5 years have been collected and described by Lucke-Wold et al. in 2016 [14].

3. Role of Thromboinflammation in aSAH

3.1. Microthrombus Formation

Within the first few days after aSAH, delayed ischemia in cerebral microcirculation contributes to early brain injury and mortality [109,110,111,112]. Evidence for an early platelet activation after aSAH has been reported in both experimental and clinical aSAH [112,113]. Intravital microscopy of cerebral microcirculation could directly demonstrate the formation of platelet microthrombi in a mouse model of aSAH and showed that thrombus formation in cerebral microcirculation contributes to blood flow deficits [114,115]. Increased endothelial expression of P-selectin and platelet deposits could also be documented by immunohistochemical and electron microscopic studies [116]. Moreover, early microclot formation in cerebral microcirculation has been reported in a rabbit model of aSAH [117]. In addition, microemboli in small cerebral arteries in patients dying within 2 days after aSAH have also been documented in autopsies [118].

3.2. The von Willebrand Factor (vWF) and ADAMTS-13

Increased activity of vWF is associated with cerebrovascular thrombosis [119]. Concomitantly, the activity of the vWF-cleaving protease ADAMTS-13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) decreases [119]. ADAMTS-13 has been described as a key protein in linking thrombosis with inflammation in injured brain [119,120,121]. Recombinant human ADAMTS-13 reduced microvascular thrombus formation and brain injury, when administered minutes after aSAH in both wildtype and ADAMTS−/− mice [122,123,124]. Interestingly, neuroinflammation, as monitored by the numbers of IBA-1 positive microglia in brain tissue, was reduced by recombinant human ADAMTS-13 and in vWF-deficient mice, whereas it was increased in ADAMTS-13 gene-deficient mice [119,123]. Plasma levels of ADAMTS-13 were reduced in the early phase after aSAH in patients with delayed cerebral ischemia [125]. In stroke patients treated with intravenous thrombolysis, low ADAMTS-13 plasma levels were associated with a worse outcome and high levels of inflammatory cytokines [126]. Thus, ADAMTS-13 has been identified as a potential biomarker for delayed cerebral ischemia after aSAH [127,128].

3.3. The Contact-Kinin System and Activated FXII

The contact-kinin pathway has been shown to play a role in the pathology of ischemic stroke and traumatic brain injury, not only by fostering vascular permeability and inflammation via kinins such as bradykinin, but also by promoting thrombus formation through the activation of the intrinsic pathway (also known as the contact pathway) [129,130]. Activated FXII (FXIIa) triggers the intrinsic coagulation cascade via the activation of FXI and induces a cascade of events by the cleavage of plasma kallikrein leading to the release of the inflammatory mediator bradykinin [129,130]. In experimental models of cerebral ischemia and traumatic brain injury, the genetic deletion of FXII or a pharmacological inhibition of FXIIa prevents microvascular thrombosis and neuroinflammation leading to reduced neuronal cell loss and improved functional neurological recovery [131,132,133,134,135,136]. Interestingly, the deficiency or pharmacologic blockade of FXII was reported to reduce neuroinflammation and render mice less susceptible to experimental autoimmune encephalomyelitis [137]. Remarkably, these beneficial effects are not accompanied by an increased risk of bleeding in experimental stroke or traumatic brain injury [131,132,135,138]. FXII deficiency actually reduced bleeding induced by tissue plasminogen activator (tPA) in experimental stroke [139]. Even if the effects of FXIIa inhibition in experimental models of aSAH have not been studied yet, therapies targeting FXIIa may also have translational potential in clinical aSAH.

3.4. Platelet Receptors

Apart from the FXIIa-mediated platelet activation, other platelet proteins contribute to inflammation after brain injuries [121,140]. Here, the platelet glycoprotein (GP)Ib-mediated platelet activation seems to be crucial for the interaction with immune cells. In experimental models of stroke, the activation of T-cell subsets contributes to brain damage in part by a platelet-dependent mechanism [141,142,143]. GPIb as a part of the GPIb-IX-V complex mediates the initial binding of platelets to the subendothelial matrix as a first step of thrombus formation. Inhibition of GPIb by neutralizing Fab fragments has anti-thrombotic and anti-inflammatory effects in mouse models of stroke [141,142,143]. Again, the beneficial effect was not associated with an increased risk of bleeding. Recent studies in mouse models of stroke have provided further evidence for T-cell interactions with platelets [144].

3.5. Potential Neuroprotective Approaches

The selective inhibition of the aforementioned pathophysiological events has been tested in experimental models of stroke, traumatic brain injury and aSAH as well as treatments targeting platelets [145,146,147,148,149], clot clearance [150], brain inflammation and immune cell infiltration [111,151,152,153,154]. These potential neuroprotective approaches are summarized in Table 2.

4. Role of Metabolism in aSAH

Similarly to the previously mentioned pathophysiological mechanisms and therapy targets, cerebral metabolism, which is already known for important pathological changes in various brain injuries such as TBI or stroke, came to the fore when a closer look was taken at aSAH pathophysiology [155]. Energy dysfunction arises in the early phase of aSAH and remains for a prolonged period of time after the event of bleeding. Therefore, research regarding the changes in post-aSAH energy metabolism inside the brain could help to understand the underlying pathophysiology of cerebral energy dysregulation in aSAH, investigating its impact on outcome and improve therapy management in clinical practice.

4.1. Metabolism with Regard to Early Brain Injury

A sudden bleeding in the subarachnoid space, commonly caused by an aneurysmal rupture, leads to a rapid increase in intracranial pressure (ICP), instantly followed by a reduction in cerebral perfusion pressure (CPP) and cerebral blood flow (CBF) [156]. While CPP recovers fast by a compensatory raise of mean artery blood pressure (MABP), CBF further decreases and stays below its normal levels for a prolonged period of time [8]. In addition to the decrease in CPP other mechanisms like ongoing acute artery vasoconstriction seem to cause cerebral hypoperfusion—a so-called low-flow-status of the brain. This sudden cerebral perfusion deficit ends up in multiple metabolic disturbances, one of a multitude of factors, which can lead to ischemic brain injury [156]. To maintain their normal function, neurons strictly depend on continuous supply with oxygen and glucose, the latter being the main source of energy for the brain. Via oxidative phosphorylation within the mitochondria, adequate amounts of adenosine triphosphate (ATP) were generated. In the case of acute cerebral ischemia, oxygen supply is disrupted and oxidative phosphorylation has to switch to insufficient anaerobic glycolysis in neurons and other brain-resident cells, leading to the accumulation of lactate in the form of an acidosis [157]. In combination with the reduced generation of ATP, this results in ion channel dysfunction, the disruption of normal cell membrane potential, production of reactive oxygen species and finally brain tissue damage by neuronal cell apoptosis [158].
Both preclinical and clinical trials repeatedly showed an increase in lactate, glutamate and lactate to the pyruvate ratio (LPR) in the interstitial brain tissue after aSAH, measured via cerebral microdialysis (CMD), as an indicator of brain metabolism derangement [8,159,160,161]. Nevertheless, the cellular level of metabolism disruption in neurons is still unknown and remains the subject of current investigations. Carpenter et al. found a significant reduction in global cerebral metabolic rate for oxygen (CMRO2) in 11 patients within the first few days after aSAH, measured via positron emission tomography (PET), independent of vasospasm-induced ischemia, hydrocephalus or intracerebral hematoma [162]. Further examinations of the course of hemodynamic parameters, CBF and tissue oxygenation (ptiO2) in an aSAH-animal model by Westermaier et al. detected an excess of tissue oxygenation several hours after aSAH, with any knowledge of prolonged increase in lactate, glutamate and LPR as metabolites of anaerobic metabolism pathways suggesting a disturbed cellular oxygen utilization and cerebral metabolic depression [163]. The basic idea is, that the affected cells are not able to use oxygen by oxidative glucose reduction via the metabolites pyruvate and acetyl-CoA, followed by the citric acid cycle and finally the respiratory chain. One or more steps of this energy metabolism pathway seem to be disturbed. In this context, Lilla et al. demonstrated for the first time a reduction in pyruvate dehydrogenase (PDH) activity following aSAH, independent of the supply of substrates [9]. As PDH is the key enzyme of the citric acid cycle, its activity reduction could be an independent factor contributing to a derangement of oxidative metabolism, a failure of oxygen utilization and secondary brain damage.
Furthermore, it is known that ischemic brain injury leads to the depolarization of the mitochondrial membrane potential, also causing a decrease in ATP production and the apoptosis of neuronal cells in the end [164]. Interestingly, Hayakawa et al. showed a release of functional mitochondria from astrocytes in the extracellular space after an experimental stroke in mice, finally entering neurons and triggering cell surviving signals [165]. Conversely, derangements in this mitochondria release pathway led to worse neurological outcome. According to that, further studies proved extracellular mitochondria in CSF both in rats and humans after aSAH, showing a significantly decreased mitochondrial membrane potential in the early after the aSAH phase [166]. Similar results were found in aSAH-patients with DCI [167]. Within the aSAH cohort, patients with higher mitochondrial membrane potential were even correlated with better neurological recovery 3 months after ictus [166]. In summary, these studies suggest that the extracellular mitochondria are a potential biomarker for the occurrence and recovery of brain injury. Due to lacking further studies, it remains unclear whether these endogenous neuroprotective mitochondrial transfer mechanisms may be exogenous therapeutical targets.

4.2. Metabolism with Regard to Delayed Brain Injury

One of the best studied mechanisms of delayed brain injury after aSAH is DCI, which commonly occurs 3–14 days after the bleeding event and represents a main cause for poor functional outcome after aSAH besides EBI. Until recently, the major cause of DCI has been thought to be CVS [168]. As vasospasm-targeted therapies in many clinical trials did not improve post-aSAH outcome, and that there were some indications that vasospasm and DCI occur separately in different locations, studies successfully looked for other DCI-underlying mechanisms [169,170]. As already mentioned, these mechanisms include, among others, cerebral vascular dysfunction, microthrombosis and neuroinflammation [171].
With this intention, experimental and clinical trials were performed to investigate the interdependency between cerebral energy metabolism and DCI. In comparison with the early phase after aSAH, similar results were received by using CMD: elevation of lactate, LPR and glutamate, while glucose and pyruvate levels were reduced in the post-aSAH long-term phase [172]. Interestingly, several studies detected significantly increased LPR and decreased glucose levels 12–16 h before DCI onset in aSAH patients, proposing these metabolites as indicators for DCI and potential help for preventing severe complications in the delayed phase of aSAH [173,174,175]. However, further studies are needed to investigate the possible treatment effects on energy metabolism and DCI as convincing results have barely existed until now. The limitations of the mentioned studies above, all using CMD as diagnostic tool, are restricted sensitivity due to the local measurement (ischemia distant to the monitoring catheter may not be detected) and restricted specificity because of the impossible differentiation between ischemic and non-ischemic metabolism derangements [172].

4.3. Potential Neuroprotective Approaches Targeting Metabolism

In the search for novel neuroprotective therapy options in aSAH, due to the illustrated mechanisms it seems that deranged cerebral metabolism could be a potential therapy target. Ca2+-channel blockers, like nimodipine, which are common part of therapy management in aSAH all over the world, were found to provide an advantage in terms of metabolic disruption, histological damage and clinical outcome after cerebral ischemia [176]. While the underlying mechanism is not cleared up yet, it may be worth having a closer look at the cellular level of metabolic disturbances. As Lilla et al. suggested, the inactivation of PDH could play a critical role in EBI after aSAH, so that preventing this inactivity may act as a neuroprotective factor [9]. Dichloroacetate (DCA) is a small molecule that crosses the BBB and stimulates PDH activity by the inhibition of PDH kinase [177]. Over the years, trials with different animal models inducing stroke or traumatic brain injury showed the neuroprotective potential of DCA, which became apparent in limiting lactate acidosis, the restoration of ATP, and improving the neurological outcome after hypoperfusion [178,179]. Kho et al. even found evidence that oxidative stress, the activation of microglia, BBB disturbance and even neuronal cell death in hypoglycemia-induced ischemia are decreased by DCA [180]. As the synopsis of all these findings, one may draw the conclusion that DCA supports metabolic recovery and therefore raises the ischemic limit of brain cells at risk of neuronal death. As DCA has not been investigated in aSAH models to date, a transfer to this disease as novel neuroprotective therapy option could be revealing.
As it is capable of the restoration of the oxidative cell metabolism, a neuroprotective effect of acetyl-L-carnitine (ALCAR) was also proven in multiple both experimental and clinical studies [181,182,183,184]. The acetylcarnitine-CoA transferase helps ALCAR entering the citric acid cycle, corresponding to a “bypass” of the PDH reaction. Therefore, the application of ALCAR could significantly lower the brain lactate level, restore ATP and even improve the neurological outcome [182,185].
In addition, mitochondrial dysfunction after aSAH is shown to activate the autophagy of neuronal cells, again one of a multitude of factors leading to EBI and DCI [44]. Therefore, targeting the autophagy–lysosomal system could be a conceivable therapy approach. This system seems to prevent cell surviving mechanisms, as long as its function is appropriate. From the moment that lysosomal-triggered autophagy gets out of control, i.e., after aSAH, an increase in the rate of cell death can be detected [186]. On this occasion worth mentioning, Chen et al. investigated the effect of epigallocatechin-3-gallate (EGCG) by an experimental study, coming to the conclusion that this active metabolite of tea catechin has the potential to lessen mitochondrial membrane potential depolarization, autophagy dysfunction and ultimately even neurological deficits [187]. Again, concerning this matter, further studies are urgently needed.

5. Role of Cerebral Vasospasm in aSAH

5.1. Pathophysiology of Cerebral Vasospasm

The exact pathophysiology of CVS occurrence is still not completely understood, while many different suggested mechanisms such as prolonged smooth muscle contraction, endothelial damage or increased endothlin-1 production exist [188].
Nevertheless, CVS demonstrates the response to damaged blood vessels due to the degradation products of red blood cells and secondary inflammation-induced vasoconstriction as the activation of the neutrophils and production of cytokines, as mentioned earlier. Moreover, different phases of CVS, including an early and a delayed response, also seem to exist [28].
In contrast to macrovascular factors, furthermore, changes in microcirculation especially seem to play a significant role in the development of delayed cerebral ischemia [189].
Endothelin 1 is known for its vasoconstrictive properties and the fact that it is overproduced in aSAH. Thus, it initially appeared to be a promising target for the treatment of CVS. Subsequently, several trials were initiated to investigate the effect of endothelin receptor antagonists. The probably most popular agent is clazosentan [190]. The phase II CONSCIOUS-1 trial demonstrated a dose-dependent reduction in CVS under medication with clazosentan [191]. The following phase III trial (CONSCIOUS-2) failed to show an improvement of the clinical outcome while the downstreamed CONSCIOUS-3 trial had to be stopped previously due to concerning side effects under therapy with clazosentan [192,193]. In summary, despite a role in reducing CVS, no clinical benefit was verified under medication with endothelin receptor antagonists.

5.2. Potential Neuroprotective Approaches Targeting Cerebral Vasospasm and Hypoperfusion

5.2.1. Magnesium Sulfate

Due to a loss of ATP and ischemic depolarization, a cellular calcium influx results after aSAH. Therefore, calcium antagonists became an interesting target in the prevention of CVS.
A deficit of serum magnesium results in increased secondary cerebral ischemia. Magnesium ameliorates rheological function and dilates blood vessels as it works as a natural calcium blocker. Hypomagnesemia in aSAH patients was found to be correlated with the amount of blood in the subarachnoid space and neurological status. When developing during medical treatment, hypomagnesemia correlates with ischemic infarctions [194].
Oral intake does not increase the serum concentrations significantly.
Several clinical studies examined the effects of magnesium treatment after aSAH with partially conflicting results [195]. One of the reservations refers to insufficiently crossing the blood–brain barrier. While several studies demonstrated a reduction in delayed cerebral ischemia and an ameliorated neurological outcome [196,197,198,199], two large trials failed to show a reduction in secondary ischemia as well as the improvement of clinical outcome [200,201], most likely dependent on the co-medication with nimodipine which works as a competitive antagonist.
Even though just one trial could show neuroprotective results, treatment with nimodipine [202], a dihydropyridine calcium antagonist, is still established worldwide.
Recent results of two clinical trials with the intracisternal application of magnesium sulfate demonstrated significantly less CVS or delayed cerebral ischemia while establishing a better functional outcome in contrast to the control groups. The additional administration of intravenous hydrogen could further show supplementary effects by finding reduced liquor levels of neuron-specific enolase, as a marker of neuronal injury, as well as reduced malondialdehyde, which is an indicator of oxidative stress [203,204].

5.2.2. Hypercapnia

After aSAH, the period of expected CVS has its maximum between day 4 and 14 due to suspended autoregulation. Under physiological conditions, the self-regulating mechanism is able to adapt to changes of arterial blood pressure for keeping the CBF constant, inter alia, by arterial partial pressure of carbon dioxide (PaCO2). Subsequently, a therapeutic use of changes in PaCO2 to increase the CBF was investigated in the clinical studies of our group [205,206]. It could be shown that within a clinical study including 12 patients with poor grad aSAH, CBF reproducibly increased during controlled phases of hypercapnia and remained raised within the first hour after downgrading to baseline without generating rebound effects resulting in a low incidence of secondary infarctions and a relatively good neurological outcome. The side effect of a mild increase in the intracranial pressure due to enhanced CBF was buffered by CSF drainage and failed to reach a pathological level according to the investigation by Petridis et al. studying the effect of permissive hypercapnia in aSAH patients [207].
The promising results of this non-pharmacological treatment will be further evaluated in a randomized multicenter trial.
In a dose optimization study of our group, temporary hypercapnia of 45 min was verified to be the optimum duration for therapeutic use (unpublished own data, manuscript submitted).

5.2.3. Hypothermia

A neuroprotective effect due to mild hypothermia was shown in a few experimental trials [208,209,210]. ICP was significantly lowered and CBF ameliorated. Even a reduced rate of injured neurons was shown, though it remains unclear whether hypothermia causes an attenuating effect or only delays brain injury. In patients with severe aSAH, therapeutic hypothermia achieved a reduction in arterial flow velocity [211]. Neuroprotective targets, potential agents and therapeutic strategies in different compartments following aSAH are summarized in Figure 1.

6. Conclusions

Aneurysmal subarachnoid hemorrhage continues to be a difficult complex cerebrovascular disease with a consistent limitation of pharmacological treatment options. Morbidity and mortality remain high despite the implementation of promising therapies such as nimodipine for treating cerebral vasospasm, new mechanisms of pathophysiology of aSAH occurred-like inflammatory processes or metabolic derangements. Having a closer look at these mechanisms, it is clear that several dysregulations take place in different compartments—vessel wall, subarachnoid space, brain parenchyma and cellular level—at different points in time—EBI/DBI, early and delayed vasospasm. Thus, several brain injury pathways must be influenced at the right place and preferably the right time to optimize therapeutic efficacy in general. While therapeutical strategies at the metabolic level are only in their early phase, no standard of care could be established yet with anti-inflammatory strategies. The non-pharmacological opportunities are promising, especially targeting vasospasm and reducing DCI for a better functional outcome. Translational clinical data should notably be in focus of future research. As beyond our scope, this review does not point out every pathophysiological aspect that is or has been under investigation in aSAH research.
Since no therapeutical breakthrough in aSAH has been made to date, and as expected further research is needed, it is vital to develop an idea of its consequences in terms of its outcome and developing potential therapies efficiently targeting brain injury.

Author Contributions

N.L. conceptualized the idea, contributed to the initial manuscript draft, reviewed and edited it. J.W.; A.B. and A.-L.S. contributed significantly to the initial manuscript draft. J.W. and A.B. designed and edited Figure 1. A.-L.S.; E.K. and T.W. critically reviewed the final draft of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This publication was supported by the Open Access Publication Fund of the University of Wuerzburg.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

aSAHAneurysmal subarachnoid hemorrhage
CVSCerebral vasospasm
DCIDelayed cerebral ischemia
BBBBlood-brainbarrier
CSFCerebrospinal fluid
TLR4Toll-like receptor 4
NADPHNicotinamide adenine dinucleotide phosphate
CNSCentral nervous system
ILInterleukin
TNFαTumor necrosis factor alpha
NF-kBNuclear factor kappa-light-chain-enhancer of activated B cells
eCBsEndocannabinoids
AEAAnandamide
2-AG2-arachidonoyl-glycerol
ECSEndocannabinoid system
CBRsCannabinoid receptors
CB1RCannabinoid type 1 receptor
CB2RCannabinoid type 2 receptor
MMPsMetalloproteinases
HMGB1High mobility group box 1
TBITraumatic brain injury
EBIEarly brain injury
DAMPDamage-associated molecular pattern
RAGEReceptor for advanced glycation end products
JAK2Janus-Kinase 2
STAT3Signal transducer and activator of transcription 3
COX-2Cyclooxygenase-2
PGE-2Prostaglandin E2
IL-1RaIL-1 receptor antagonist
vWFvon Willebrand Factor
ADAMTS-13A disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13
tPATissue plasminogen activator+
GPPlatelet glycoprotein
ICHIntracerebral hemorrhage
ICPIntracranial pressure
CPPCerebral perfusion pressure
CBFCerebral blood flow
MABPMean artery blood pressure
ATPAdenosine triphosphate
LPRLactate-to-pyruvate ratio
CMDCerebral microdialysis
CMRO2Cerebral metabolic rate for oxygen
PETPositron emission tomography
ptiO2Tissue oxygenation
PDHPyruvate dehydrogenase
DCADichloroacetate
ALCARAcetyl-L-carnitine
EGCG Epigallocatechin-3-gallate
PaCO2Arterial partial pressure of carbon dioxide

References

  1. Hackenberg, K.A.M.; Hanggi, D.; Etminan, N. Unruptured intracranial aneurysms. Stroke J. Cereb. Circ. 2018, 49, 2268–2275. [Google Scholar] [CrossRef] [PubMed]
  2. Macdonald, R.L. Delayed neurological deterioration after subarachnoid haemorrhage. Nat. Rev. Neurol. 2014, 10, 44–58. [Google Scholar] [CrossRef]
  3. Schneider, U.C.; Xu, R.; Vajkoczy, P. Inflammatory events following subarachnoid hemorrhage (SAH). Curr. Neuropharmacol. 2018, 16, 1385–1395. [Google Scholar] [CrossRef]
  4. Vergouwen, M.D.; Vermeulen, M.; van Gijn, J.; Rinkel, G.J.; Wijdicks, E.F.; Muizelaar, J.P.; Mendelow, A.D.; Juvela, S.; Yonas, H.; Terbrugge, K.G.; et al. Definition of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage as an outcome event in clinical trials and observational studies: Proposal of a multidisciplinary research group. Stroke J. Cereb. Circ. 2010, 41, 2391–2395. [Google Scholar] [CrossRef] [Green Version]
  5. Jaja, B.N.R.; Saposnik, G.; Lingsma, H.F.; Macdonald, E.; Thorpe, K.E.; Mamdani, M.; Steyerberg, E.W.; Molyneux, A.; Manoel, A.L.O.; Schatlo, B.; et al. Development and validation of outcome prediction models for aneurysmal subarachnoid haemorrhage: The SAHIT multinational cohort study. BMJ 2018, 360, 5745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Geraghty, J.R.; Davis, J.L.; Testai, F.D. Neuroinflammation and microvascular dysfunction after experimental subarachnoid hemorrhage: Emerging components of early brain injury related to outcome. Neurocritical. Care 2019, 31, 373–389. [Google Scholar] [CrossRef] [PubMed]
  7. Sarrafzadeh, A.; Haux, D.; Sakowitz, O.; Benndorf, G.; Herzog, H.; Kuechler, I.; Unterberg, A. Acute focal neurological deficits in aneurysmal subarachnoid hemorrhage: Relation of clinical course, CT findings, and metabolite abnormalities monitored with bedside microdialysis. Stroke J. Cereb. Circ. 2003, 34, 1382–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Westermaier, T.; Jauss, A.; Eriskat, J.; Kunze, E.; Roosen, K. The temporal profile of cerebral blood flow and tissue metabolites indicates sustained metabolic depression after experimental subarachnoid hemorrhage in rats. Neurosurgery 2011, 68, 223–229, discussion 229–230. [Google Scholar] [CrossRef]
  9. Lilla, N.; Fullgraf, H.; Stetter, C.; Kohler, S.; Ernestus, R.I.; Westermaier, T. First description of reduced pyruvate dehydrogenase enzyme activity following subarachnoid hemorrhage (SAH). Front. Neurosci. 2017, 11, 37. [Google Scholar] [CrossRef] [PubMed]
  10. Provencio, J.J. Inflammation in subarachnoid hemorrhage and delayed deterioration associated with vasospasm: A review. Acta Neurochir. Suppl. 2013, 115, 233–238. [Google Scholar]
  11. Sela, S.; Shurtz-Swirski, R.; Awad, J.; Shapiro, G.; Nasser, L.; Shasha, S.M.; Kristal, B. The involvement of peripheral polymorphonuclear leukocytes in the oxidative stress and inflammation among cigarette smokers. Isr. Med. Assoc. J. 2002, 4, 1015–1019. [Google Scholar]
  12. Zeyu, Z.; Yuanjian, F.; Cameron, L.; Sheng, C. The role of immune inflammation in aneurysmal subarachnoid hemorrhage. Exp. Neurol. 2021, 336, 113535. [Google Scholar] [CrossRef] [PubMed]
  13. Provencio, J.J.; Fu, X.; Siu, A.; Rasmussen, P.A.; Hazen, S.L.; Ransohoff, R.M. CSF neutrophils are implicated in the development of vasospasm in subarachnoid hemorrhage. Neurocrit. Care 2010, 12, 244–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lucke-Wold, B.P.; Logsdon, A.F.; Manoranjan, B.; Turner, R.C.; McConnell, E.; Vates, G.E.; Huber, J.D.; Rosen, C.L.; Simard, J.M. Aneurysmal subarachnoid hemorrhage and neuroinflammation: A comprehensive review. Int. J. Mol. Sci. 2016, 17, 497. [Google Scholar] [CrossRef] [PubMed]
  15. Schneider, U.C.; Schiffler, J.; Hakiy, N.; Horn, P.; Vajkoczy, P. Functional analysis of Pro-inflammatory properties within the cerebrospinal fluid after subarachnoid hemorrhage in vivo and in vitro. J. Neuroinflamm. 2012, 9, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59. [Google Scholar] [CrossRef]
  17. Banks, W.A.; Kastin, A.J.; Broadwell, R.D. Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation 1995, 2, 241–248. [Google Scholar] [CrossRef]
  18. Bulters, D.; Gaastra, B.; Zolnourian, A.; Alexander, S.; Ren, D.; Blackburn, S.L.; Borsody, M.; Dore, S.; Galea, J.; Iihara, K.; et al. Haemoglobin scavenging in intracranial bleeding: Biology and clinical implications. Nat. Rev. Neurol. 2018, 14, 416–432. [Google Scholar] [CrossRef]
  19. Macdonald, R.L.; Marton, L.S.; Andrus, P.K.; Hall, E.D.; Johns, L.; Sajdak, M. Time course of production of hydroxyl free radical after subarachnoid hemorrhage in dogs. Life Sci. 2004, 75, 979–989. [Google Scholar] [CrossRef]
  20. Gallia, G.L.; Tamargo, R.J. Leukocyte-endothelial cell interactions in chronic vasospasm after subarachnoid hemorrhage. Neurol. Res. 2006, 28, 750–758. [Google Scholar] [CrossRef]
  21. Dietrich, H.H.; Dacey, R.G., Jr. Molecular keys to the problems of cerebral vasospasm. Neurosurgery 2000, 46, 517–530. [Google Scholar] [CrossRef]
  22. Klein, R.S.; Garber, C.; Howard, N. Infectious immunity in the central nervous system and brain function. Nat. Immunol. 2017, 18, 132–141. [Google Scholar] [CrossRef]
  23. Mathiesen, T.; Andersson, B.; Loftenius, A.; von Holst, H. Increased interleukin-6 levels in cerebrospinal fluid following subarachnoid hemorrhage. J. Neurosurg. 1993, 78, 562–567. [Google Scholar] [CrossRef] [Green Version]
  24. Kikuchi, T.; Okuda, Y.; Kaito, N.; Abe, T. Cytokine production in cerebrospinal fluid after subarachnoid haemorrhage. Neurol. Res. 1995, 17, 106–108. [Google Scholar] [CrossRef]
  25. Figueiredo, R.T.; Fernandez, P.L.; Mourao-Sa, D.S.; Porto, B.N.; Dutra, F.F.; Alves, L.S.; Oliveira, M.F.; Oliveira, P.L.; Graça-Souza, A.V.; Bozza, M.T. Characterization of heme as activator of Toll-like receptor 4. J. Biol. Chem. 2007, 282, 20221–20229. [Google Scholar] [CrossRef] [Green Version]
  26. Crowley, R.W.; Medel, R.; Kassell, N.F.; Dumont, A.S. New insights into the causes and therapy of cerebral vasospasm following subarachnoid hemorrhage. Drug. Discov. Today 2008, 13, 254–260. [Google Scholar] [CrossRef]
  27. Pradilla, G.; Chaichana, K.L.; Hoang, S.; Huang, J.; Tamargo, R.J. Inflammation and cerebral vasospasm after subarachnoid hemorrhage. Neurosurg. Clin. N. Am. 2010, 21, 365–379. [Google Scholar] [CrossRef]
  28. Hanafy, K.A. The role of microglia and the TLR4 pathway in neuronal apoptosis and vasospasm after subarachnoid hemorrhage. J. Neuroinflamm. 2013, 10, 83. [Google Scholar] [CrossRef] [Green Version]
  29. Heinz, R.; Brandenburg, S.; Nieminen-Kelhä, M.; Kremenetskaia, I.; Boehm-Sturm, P.; Vajkoczy, P.; Schneider, U.C. Microglia as target for anti-inflammatory approaches to prevent secondary brain injury after subarachnoid hemorrhage (SAH). J. Neuroinflamm. 2021, 18, 36. [Google Scholar] [CrossRef] [PubMed]
  30. Schneider, U.C.; Davids, A.M.; Brandenburg, S.; Müller, A.; Elke, A.; Magrini, S.; Atangana, E.; Turkowski, K.; Finger, T.; Gutenberg, A.; et al. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathol. 2015, 130, 215–231. [Google Scholar] [CrossRef]
  31. Patricio, F.; Morales-Andrade, A.A.; Patricio-Martinez, A.; Limon, I.D. Cannabidiol as a therapeutic target: Evidence of its neuroprotective and neuromodulatory function in parkinson’s disease. Front. Pharm. 2020, 11, 595635. [Google Scholar] [CrossRef] [PubMed]
  32. Benyo, Z.; Ruisanchez, E.; Leszl-Ishiguro, M.; Sandor, P.; Pacher, P. Endocannabinoids in cerebrovascular regulation. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H785–H801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Duffy, S.S.; Hayes, J.P.; Fiore, N.T.; Moalem-Taylor, G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021, 190, 108555. [Google Scholar] [CrossRef]
  34. Zhang, X.; Ares, W.J.; Taussky, P.; Ducruet, A.F.; Grandhi, R. Role of matrix metalloproteinases in the pathogenesis of intracranial aneurysms. Neurosurg. Focus 2019, 47, E4. [Google Scholar] [CrossRef] [Green Version]
  35. Chaudhry, S.R.; Hafez, A.; Rezai Jahromi, B.; Kinfe, T.M.; Lamprecht, A.; Niemela, M.; Muhammad, S. Role of damage associated molecular pattern molecules (DAMPs) in aneurysmal subarachnoid hemorrhage (aSAH). Int. J. Mol. Sci. 2018, 19, 2035. [Google Scholar] [CrossRef] [Green Version]
  36. Balanca, B.; Desmurs, L.; Grelier, J.; Perret-Liaudet, A.; Lukaszewicz, A.C. DAMPs and RAGE pathophysiology at the acute phase of brain injury: An overview. Int. J. Mol. Sci. 2021, 22, 2439. [Google Scholar] [CrossRef]
  37. Aucott, H.; Lundberg, J.; Salo, H.; Klevenvall, L.; Damberg, P.; Ottosson, L.; Andersson, U.; Holmin, S.; Erlandsson Harris, H. Neuroinflammation in response to intracerebral injections of different HMGB1 redox isoforms. J. Innate Immun. 2018, 10, 215–227. [Google Scholar] [CrossRef] [PubMed]
  38. Haruma, J.; Teshigawara, K.; Hishikawa, T.; Wang, D.; Liu, K.; Wake, H.; Mori, S.; Takahashi, H.K.; Sugiu, K.; Date, I.; et al. Anti-high mobility group box-1 (HMGB1) antibody attenuates delayed cerebral vasospasm and brain injury after subarachnoid hemorrhage in rats. Sci. Rep. 2016, 6, 37755. [Google Scholar] [CrossRef]
  39. Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Othman, I.; Shaikh, M.F. HMGB1-Mediated neuroinflammatory responses in brain injuries: Potential mechanisms and therapeutic opportunities. Int. J. Mol. Sci. 2020, 21, 4609. [Google Scholar] [CrossRef]
  40. Sun, Q.; Wu, W.; Hu, Y.C.; Li, H.; Zhang, D.; Li, S.; Li, W.; Li, W.D.; Ma, B.; Zhu, J.H.; et al. Early release of high-mobility group box 1 (HMGB1) from neurons in experimental subarachnoid hemorrhage in vivo and in vitro. J. Neuroinflamm. 2014, 11, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. An, J.Y.; Pang, H.G.; Huang, T.Q.; Song, J.N.; Li, D.D.; Zhao, Y.L.; Ma, X.D. AG490 ameliorates early brain injury via inhibition of JAK2/STAT3-mediated regulation of HMGB1 in subarachnoid hemorrhage. Exp. Med. 2018, 15, 1330–1338. [Google Scholar] [CrossRef] [Green Version]
  42. Gu, X.J.; Xu, J.; Ma, B.Y.; Chen, G.; Gu, P.Y.; Wei, D.; Hu, W.X. Effect of glycyrrhizin on traumatic brain injury in rats and its mechanism. Chin. J. Traumatol. 2014, 17, 1–7. [Google Scholar]
  43. Chaudhry, S.R.; Guresir, A.; Stoffel-Wagner, B.; Fimmers, R.; Kinfe, T.M.; Dietrich, D.; Lamprecht, A.; Vatter, H.; Guresir, E.; Muhammad, S. Systemic high-mobility group box-1: A novel predictive biomarker for cerebral vasospasm in aneurysmal subarachnoid hemorrhage. Crit. Care Med. 2018, 46, e1023–e1028. [Google Scholar] [CrossRef]
  44. Pawlowska, E.; Szczepanska, J.; Wisniewski, K.; Tokarz, P.; Jaskolski, D.J.; Blasiak, J. NF-kappaB-mediated inflammation in the pathogenesis of intracranial aneurysm and subarachnoid hemorrhage. does autophagy play a role? Int. J. Mol. Sci. 2018, 19, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Frosen, J.; Cebral, J.; Robertson, A.M.; Aoki, T. Flow-induced, inflammation-mediated arterial wall remodeling in the formation and progression of intracranial aneurysms. Neurosurg. Focus 2019, 47, E21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sen, R.; Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986, 46, 705–716. [Google Scholar] [CrossRef]
  47. Perkins, N.D. Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat. Rev. Mol. Cell Biol. 2007, 8, 49–62. [Google Scholar] [CrossRef]
  48. Aoki, T.; Frosen, J.; Fukuda, M.; Bando, K.; Shioi, G.; Tsuji, K.; Ollikainen, E.; Nozaki, K.; Laakkonen, J.; Narumiya, S. Prostaglandin E2-EP2-NF-kappaB signaling in macrophages as a potential therapeutic target for intracranial aneurysms. Sci. Signal. 2017, 10, eaah6037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Liu, Y.; Cai, H.; Wang, Z.; Li, J.; Wang, K.; Yu, Z.; Chen, G. Induction of autophagy by cystatin C: A potential mechanism for prevention of cerebral vasospasm after experimental subarachnoid hemorrhage. Eur. J. Med. Res. 2013, 18, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Yan, F.; Li, J.; Chen, J.; Hu, Q.; Gu, C.; Lin, W.; Chen, G. Endoplasmic reticulum stress is associated with neuroprotection against apoptosis via autophagy activation in a rat model of subarachnoid hemorrhage. Neurosci. Lett. 2014, 563, 160–165. [Google Scholar] [CrossRef]
  51. Luo, C.; Yao, X.; Li, J.; He, B.; Liu, Q.; Ren, H.; Liang, F.; Li, M.; Lin, H.; Peng, J.; et al. Paravascular pathways contribute to vasculitis and neuroinflammation after subarachnoid hemorrhage independently of glymphatic control. Cell Death Dis. 2016, 7, e2160. [Google Scholar] [CrossRef] [PubMed]
  52. Mestre, H.; Kostrikov, S.; Mehta, R.I.; Nedergaard, M. Perivascular spaces, glymphatic dysfunction, and small vessel disease. Clin. Sci. 2017, 131, 2257–2274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191. [Google Scholar] [CrossRef] [PubMed]
  54. Louveau, A.; Herz, J.; Alme, M.N.; Salvador, A.F.; Dong, M.Q.; Viar, K.E.; Herod, S.G.; Knopp, J.; Setliff, J.C.; Lupi, A.L.; et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 2018, 21, 1380–1391. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, J.; Wang, L.; Xu, H.; Xing, L.; Zhuang, Z.; Zheng, Y.; Li, X.; Wang, C.; Chen, S.; Guo, Z.; et al. Meningeal lymphatics clear erythrocytes that arise from subarachnoid hemorrhage. Nat. Commun. 2020, 11, 3159. [Google Scholar] [CrossRef] [PubMed]
  56. Vadokas, G.; Koehler, S.; Weiland, J.; Lilla, N.; Stetter, C.; Westermaier, T. Early antiinflammatory therapy attenuates brain damage after sah in rats. Transl. Neurosci. 2019, 10, 104–111. [Google Scholar] [CrossRef]
  57. Gomis, P.; Graftieaux, J.P.; Sercombe, R.; Hettler, D.; Scherpereel, B.; Rousseaux, P. Randomized, double-blind, placebo-controlled, pilot trial of high-dose methylprednisolone in aneurysmal subarachnoid hemorrhage. J. Neurosurg. 2010, 112, 681–688. [Google Scholar] [CrossRef]
  58. Feigin, V.L.; Anderson, N.; Rinkel, G.J.; Algra, A.; van Gijn, J.; Bennett, D.A. Corticosteroids for aneurysmal subarachnoid haemorrhage and primary intracerebral haemorrhage. Cochrane Database Syst. Rev. 2005, 3, Cd004583. [Google Scholar] [CrossRef]
  59. Belayev, L.; Saul, I.; Huh, P.W.; Finotti, N.; Zhao, W.; Busto, R.; Ginsberg, M.D. Neuroprotective effect of high-dose albumin therapy against global ischemic brain injury in rats. Brain Res. 1999, 845, 107–111. [Google Scholar] [CrossRef]
  60. Liu, Y.; Belayev, L.; Zhao, W.; Busto, R.; Belayev, A.; Ginsberg, M.D. Neuroprotective effect of treatment with human albumin in permanent focal cerebral ischemia: Histopathology and cortical perfusion studies. Eur. J. Pharm. 2001, 428, 193–201. [Google Scholar] [CrossRef]
  61. Suarez, J.I.; Martin, R.H.; Calvillo, E.; Dillon, C.; Bershad, E.M.; Macdonald, R.L.; Wong, J.; Harbaugh, R. The Albumin in Subarachnoid Hemorrhage (ALISAH) multicenter pilot clinical trial: Safety and neurologic outcomes. Stroke 2012, 43, 683–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Singh, N.; Hopkins, S.J.; Hulme, S.; Galea, J.P.; Hoadley, M.; Vail, A.; Hutchinson, P.J.; Grainger, S.; Rothwell, N.J.; King, A.T.; et al. The effect of intravenous interleukin-1 receptor antagonist on inflammatory mediators in cerebrospinal fluid after subarachnoid haemorrhage: A phase II randomised controlled trial. J. Neuroinflamm. 2014, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Galea, J.; Ogungbenro, K.; Hulme, S.; Patel, H.; Scarth, S.; Hoadley, M.; Illingworth, K.; McMahon, C.J.; Tzerakis, N.; King, A.T.; et al. Reduction of inflammation after administration of interleukin-1 receptor antagonist following aneurysmal subarachnoid hemorrhage: Results of the Subcutaneous Interleukin-1Ra in SAH (SCIL-SAH) study. J. Neurosurg. 2018, 128, 515–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chaudhry, S.R.; Kahlert, U.D.; Kinfe, T.M.; Lamprecht, A.; Niemela, M.; Hanggi, D.; Muhammad, S. Elevated systemic IL-10 levels indicate immunodepression leading to nosocomial infections after aneurysmal subarachnoid hemorrhage (SAH) in patients. Int. J. Mol. Sci. 2020, 21, 1569. [Google Scholar] [CrossRef] [Green Version]
  65. Chaudhry, S.R.; Kinfe, T.M.; Lamprecht, A.; Niemela, M.; Dobreva, G.; Hanggi, D.; Muhammad, S. Elevated level of cerebrospinal fluid and systemic chemokine CCL5 is a predictive biomarker of clinical outcome after aneurysmal subarachnoid hemorrhage (aSAH). Cytokine 2020, 133, 155142. [Google Scholar] [CrossRef] [PubMed]
  66. Fujii, M.; Sherchan, P.; Krafft, P.R.; Rolland, W.B.; Soejima, Y.; Zhang, J.H. Cannabinoid type 2 receptor stimulation attenuates brain edema by reducing cerebral leukocyte infiltration following subarachnoid hemorrhage in rats. J. Neurol. Sci. 2014, 342, 101–106. [Google Scholar] [CrossRef] [Green Version]
  67. Fujii, M.; Sherchan, P.; Soejima, Y.; Hasegawa, Y.; Flores, J.; Doycheva, D.; Zhang, J.H. Cannabinoid receptor type 2 agonist attenuates apoptosis by activation of phosphorylated CREB-Bcl-2 pathway after subarachnoid hemorrhage in rats. Exp. Neurol. 2014, 261, 396–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Fujii, M.; Sherchan, P.; Soejima, Y.; Doycheva, D.; Zhao, D.; Zhang, J.H. Cannabinoid receptor Type 2 agonist attenuates acute neurogenic pulmonary edema by preventing neutrophil migration after subarachnoid hemorrhage in rats. Acta Neurochir. Suppl. 2016, 121, 135–139. [Google Scholar]
  69. Behrouz, R.; Birnbaum, L.; Grandhi, R.; Johnson, J.; Misra, V.; Palacio, S.; Seifi, A.; Topel, C.; Garvin, R.; Caron, J.L. Cannabis use and outcomes in patients with aneurysmal subarachnoid hemorrhage. Stroke J. Cereb. Circ. 2016, 47, 1371–1373. [Google Scholar] [CrossRef] [Green Version]
  70. De Oliveira Manoel, A.L.; Macdonald, R.L. Neuroinflammation as a target for intervention in subarachnoid hemorrhage. Front. Neurol. 2018, 9, 292. [Google Scholar] [CrossRef] [Green Version]
  71. Dong, G.; Li, C.; Hu, Q.; Wang, Y.; Sun, J.; Gao, F.; Yang, M.; Sun, B.; Mao, L. Low-Dose IL-2 treatment affords protection against subarachnoid hemorrhage injury by expanding peripheral regulatory T cells. ACS Chem. Neurosci. 2021, 12, 430–440. [Google Scholar] [CrossRef]
  72. Han, M.; Cao, Y.; Guo, X.; Chu, X.; Li, T.; Xue, H.; Xin, D.; Yuan, L.; Ke, H.; Li, G.; et al. Mesenchymal stem cell-derived extracellular vesicles promote microglial M2 polarization after subarachnoid hemorrhage in rats and involve the AMPK/NF-kappaB signaling pathway. Biomed. Pharm. 2021, 133, 111048. [Google Scholar] [CrossRef]
  73. Li, P.; Li, X.; Deng, P.; Wang, D.; Bai, X.; Li, Y.; Luo, C.; Belguise, K.; Wang, X.; Wei, X.; et al. Activation of adenosine A3 receptor reduces early brain injury by alleviating neuroinflammation after subarachnoid hemorrhage in elderly rats. Aging (Albany Ny) 2020, 13, 694–713. [Google Scholar] [CrossRef]
  74. Gao, Y.Y.; Tao, T.; Wu, D.; Zhuang, Z.; Lu, Y.; Wu, L.Y.; Liu, G.J.; Zhou, Y.; Zhang, D.D.; Wang, H.; et al. MFG-E8 attenuates inflammation in subarachnoid hemorrhage by driving microglial M2 polarization. Exp. Neurol. 2021, 336, 113532. [Google Scholar] [CrossRef]
  75. Kremer, B.; Coburn, M.; Weinandy, A.; Nolte, K.; Clusmann, H.; Veldeman, M.; Hollig, A. Argon treatment after experimental subarachnoid hemorrhage: Evaluation of microglial activation and neuronal survival as a subanalysis of a randomized controlled animal trial. Med. Gas. Res. 2020, 10, 103–109. [Google Scholar] [CrossRef]
  76. Luo, Y.; Fang, Y.; Kang, R.; Lenahan, C.; Gamdzyk, M.; Zhang, Z.; Okada, T.; Tang, J.; Chen, S.; Zhang, J.H. Inhibition of EZH2 (Enhancer of Zeste Homolog 2) Attenuates Neuroinflammation via H3k27me3/SOCS3/TRAF6/NF-kappaB (Trimethylation of Histone 3 Lysine 27/Suppressor of Cytokine Signaling 3/Tumor Necrosis Factor Receptor Family 6/Nuclear Factor-kappaB) in a Rat Model of Subarachnoid Hemorrhage. Stroke 2020, 51, 3320–3331. [Google Scholar]
  77. Chen, T.; Zhu, J.; Wang, Y.H. RNF216 mediates neuronal injury following experimental subarachnoid hemorrhage through the Arc/Arg3.1-AMPAR pathway. Faseb J. 2020, 34, 15080–15092. [Google Scholar] [CrossRef]
  78. Xu, P.; Hong, Y.; Xie, Y.; Yuan, K.; Li, J.; Sun, R.; Zhang, X.; Shi, X.; Li, R.; Wu, J.; et al. TREM-1 Exacerbates Neuroinflammatory Injury via NLRP3 Inflammasome-Mediated Pyroptosis in Experimental Subarachnoid Hemorrhage. Transl. Stroke Res. 2020. [Google Scholar] [CrossRef]
  79. Liu, G.J.; Tao, T.; Wang, H.; Zhou, Y.; Gao, X.; Gao, Y.Y.; Hang, C.H.; Li, W. Functions of resolvin D1-ALX/FPR2 receptor interaction in the hemoglobin-induced microglial inflammatory response and neuronal injury. J. Neuroinflamm. 2020, 17, 239. [Google Scholar] [CrossRef]
  80. Duan, H.; Li, L.; Shen, S.; Ma, Y.; Yin, X.; Liu, Z.; Yuan, C.; Wang, Y.; Zhang, J. Hydrogen Sulfide Reduces Cognitive Impairment in Rats After Subarachnoid Hemorrhage by Ameliorating Neuroinflammation Mediated by the TLR4/NF-kappaB Pathway in Microglia. Front. Cell Neurosci. 2020, 14, 210. [Google Scholar] [CrossRef]
  81. Liao, L.S.; Zhang, M.W.; Gu, Y.J.; Sun, X.C. Targeting CCL20 inhibits subarachnoid hemorrhage-related neuroinflammation in mice. Aging (Albany Ny) 2020, 12, 14849–14862. [Google Scholar] [CrossRef] [PubMed]
  82. Zhou, J.; Zhang, X.; Peng, J.; Xie, Y.; Du, F.; Guo, K.; Feng, Y.; Zhang, L.; Chen, L.; Jiang, Y. TSPO ligand Ro5–4864 modulates microglia/macrophages polarization after subarachnoid hemorrhage in mice. Neurosci. Lett. 2020, 729, 134977. [Google Scholar] [CrossRef] [PubMed]
  83. Peng, W.; Wu, X.; Feng, D.; Zhang, Y.; Chen, X.; Ma, C.; Shen, H.; Li, X.; Li, H.; Zhang, J.; et al. Cerebral cavernous malformation 3 relieves subarachnoid hemorrhage-induced neuroinflammation in rats through inhibiting NF-kB signaling pathway. Brain Res. Bull. 2020, 160, 74–84. [Google Scholar] [CrossRef] [PubMed]
  84. Lai, N.; Wu, D.; Liang, T.; Pan, P.; Yuan, G.; Li, X.; Li, H.; Shen, H.; Wang, Z.; Chen, G. Systemic exosomal miR-193b-3p delivery attenuates neuroinflammation in early brain injury after subarachnoid hemorrhage in mice. J. Neuroinflamm. 2020, 17, 74. [Google Scholar] [CrossRef] [PubMed]
  85. Gao, Y.; Zhuang, Z.; Lu, Y.; Tao, T.; Zhou, Y.; Liu, G.; Wang, H.; Zhang, D.; Wu, L.; Dai, H.; et al. Curcumin Mitigates Neuro-Inflammation by Modulating Microglia Polarization Through Inhibiting TLR4 Axis Signaling Pathway Following Experimental Subarachnoid Hemorrhage. Front. Neurosci. 2019, 13, 1223. [Google Scholar] [CrossRef] [PubMed]
  86. Tao, T.; Liu, G.J.; Shi, X.; Zhou, Y.; Lu, Y.; Gao, Y.Y.; Zhang, X.S.; Wang, H.; Wu, L.Y.; Chen, C.L.; et al. DHEA Attenuates Microglial Activation via Induction of JMJD3 in Experimental Subarachnoid Haemorrhage. J. Neuroinflamm. 2019, 16, 243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Xu, W.; Li, T.; Gao, L.; Zheng, J.; Yan, J.; Zhang, J.; Shao, A. Apelin-13/APJ system attenuates early brain injury via suppression of endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation and oxidative stress in a AMPK-dependent manner after subarachnoid hemorrhage in rats. J. Neuroinflamm. 2019, 16, 247. [Google Scholar] [CrossRef]
  88. Xu, W.; Mo, J.; Ocak, U.; Travis, Z.D.; Enkhjargal, B.; Zhang, T.; Wu, P.; Peng, J.; Li, T.; Zuo, Y.; et al. Activation of Melanocortin 1 Receptor Attenuates Early Brain Injury in a Rat Model of Subarachnoid Hemorrhage viathe Suppression of Neuroinflammation through AMPK/TBK1/NF-kappaB Pathway in Rats. Neurotherapeutics 2020, 17, 294–308. [Google Scholar] [CrossRef]
  89. Okada, T.; Lei, L.; Nishikawa, H.; Nakano, F.; Nakatsuka, Y.; Suzuki, H. TAK-242, Toll-Like Receptor 4 Antagonist, Attenuates Brain Edema in Subarachnoid Hemorrhage Mice. Acta Neurochir. Suppl. 2020, 127, 77–81. [Google Scholar]
  90. Zuo, Y.; Huang, L.; Enkhjargal, B.; Xu, W.; Umut, O.; Travis, Z.D.; Zhang, G.; Tang, J.; Liu, F.; Zhang, J.H. Activation of retinoid X receptor by bexarotene attenuates neuroinflammation via PPARgamma/SIRT6/FoxO3a pathway after subarachnoid hemorrhage in rats. J. Neuroinflamm. 2019, 16, 47. [Google Scholar] [CrossRef] [Green Version]
  91. Li, R.; Venkat, P.; Chopp, M.; Zhang, Q.; Yan, T.; Chen, J. RP001 hydrochloride improves neurological outcome after subarachnoid hemorrhage. J. Neurol. Sci. 2019, 399, 6–14. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, W.; Li, R.; Yin, J.; Guo, S.; Chen, Y.; Fan, H.; Li, G.; Li, Z.; Li, X.; Zhang, X.; et al. Mesenchymal stem cells alleviate the early brain injury of subarachnoid hemorrhage partly by suppression of Notch1-dependent neuroinflammation: Involvement of Botch. J. Neuroinflamm. 2019, 16, 8. [Google Scholar] [CrossRef]
  93. Liu, F.Y.; Cai, J.; Wang, C.; Ruan, W.; Guan, G.P.; Pan, H.Z.; Li, J.R.; Qian, C.; Chen, J.S.; Wang, L.; et al. Fluoxetine attenuates neuroinflammation in early brain injury after subarachnoid hemorrhage: A possible role for the regulation of TLR4/MyD88/NF-kappaB signaling pathway. J. Neuroinflamm. 2018, 15, 347. [Google Scholar] [CrossRef] [PubMed]
  94. Pang, J.; Peng, J.; Matei, N.; Yang, P.; Kuai, L.; Wu, Y.; Chen, L.; Vitek, M.P.; Li, F.; Sun, X.; et al. Apolipoprotein E Exerts a Whole-Brain Protective Property by Promoting M1? Microglia Quiescence After Experimental Subarachnoid Hemorrhage in Mice. Transl Stroke Res. 2018, 9, 654–668. [Google Scholar] [CrossRef] [PubMed]
  95. Li, R.; Liu, W.; Yin, J.; Chen, Y.; Guo, S.; Fan, H.; Li, X.; Zhang, X.; He, X.; Duan, C. TSG-6 attenuates inflammation-induced brain injury via modulation of microglial polarization in SAH rats through the SOCS3/STAT3 pathway. J. Neuroinflamm. 2018, 15, 231. [Google Scholar] [CrossRef]
  96. Xu, Z.; Shi, W.H.; Xu, L.B.; Shao, M.F.; Chen, Z.P.; Zhu, G.C.; Hou, Q. Resident Microglia Activate before Peripheral Monocyte Infiltration and p75NTR Blockade Reduces Microglial Activation and Early Brain Injury after Subarachnoid Hemorrhage. ACS Chem. Neurosci. 2019, 10, 412–423. [Google Scholar] [CrossRef]
  97. Qu, J.; Zhao, H.; Li, Q.; Pan, P.; Ma, K.; Liu, X.; Feng, H.; Chen, Y. MST1 Suppression Reduces Early Brain Injury by Inhibiting the NF-kappaB/MMP-9 Pathway after Subarachnoid Hemorrhage in Mice. Behav. Neurol. 2018, 2018, 6470957. [Google Scholar] [CrossRef] [Green Version]
  98. Zhu, Q.; Enkhjargal, B.; Huang, L.; Zhang, T.; Sun, C.; Xie, Z.; Wu, P.; Mo, J.; Tang, J.; Xie, Z.; et al. Aggf1 attenuates neuroinflammation and BBB disruption via PI3K/Akt/NF-kappaB pathway after subarachnoid hemorrhage in rats. J. Neuroinflamm. 2018, 15, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Okada, T.; Kawakita, F.; Nishikawa, H.; Nakano, F.; Liu, L.; Suzuki, H. Selective Toll-Like Receptor 4 Antagonists Prevent Acute Blood-Brain Barrier Disruption After Subarachnoid Hemorrhage in Mice. Mol. Neurobiol. 2019, 56, 976–985. [Google Scholar] [CrossRef]
  100. Tu, L.; Yang, X.L.; Zhang, Q.; Wang, Q.; Tian, T.; Liu, D.; Qu, X.; Tian, J.Y. Bexarotene attenuates early brain injury via inhibiting micoglia activation through PPARgamma after experimental subarachnoid hemorrhage. Neurol. Res. 2018, 40, 702–708. [Google Scholar]
  101. Yin, J.; Li, R.; Liu, W.; Chen, Y.; Zhang, X.; Li, X.; He, X.; Duan, C. Neuroprotective Effect of Protein Phosphatase 2A/Tristetraprolin Following Subarachnoid Hemorrhage in Rats. Front. Neurosci. 2018, 12, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Peng, Y.; Jin, J.; Fan, L.; Xu, H.; He, P.; Li, J.; Chen, T.; Ruan, W.; Chen, G. Rolipram Attenuates Early Brain Injury Following Experimental Subarachnoid Hemorrhage in Rats: Possibly via Regulating the SIRT1/NF-kappaB Pathway. Neurochem. Res. 2018, 43, 785–795. [Google Scholar] [CrossRef] [PubMed]
  103. Xie, Z.; Huang, L.; Enkhjargal, B.; Reis, C.; Wan, W.; Tang, J.; Cheng, Y.; Zhang, J.H. Recombinant Netrin-1 binding UNC5B receptor attenuates neuroinflammation and brain injury via PPARgamma/NFkappaB signaling pathway after subarachnoid hemorrhage in rats. Brain Behav. Immun. 2018, 69, 190–202. [Google Scholar] [CrossRef] [PubMed]
  104. Li, J.R.; Xu, H.Z.; Nie, S.; Peng, Y.C.; Fan, L.F.; Wang, Z.J.; Wu, C.; Yan, F.; Chen, J.Y.; Gu, C.; et al. Fluoxetine-enhanced autophagy ameliorates early brain injury via inhibition of NLRP3 inflammasome activation following subrachnoid hemorrhage in rats. J. Neuroinflamm. 2017, 14, 186. [Google Scholar] [CrossRef]
  105. Xu, H.; Li, J.; Wang, Z.; Feng, M.; Shen, Y.; Cao, S.; Li, T.; Peng, Y.; Fan, L.; Chen, J.; et al. Methylene blue attenuates neuroinflammation after subarachnoid hemorrhage in rats through the Akt/GSK-3beta/MEF2D signaling pathway. Brain Behav. Immun. 2017, 65, 125–139. [Google Scholar] [CrossRef]
  106. Xu, J.; Xu, Z.; Yan, A. Prostaglandin E2 EP4 Receptor Activation Attenuates Neuroinflammation and Early Brain Injury Induced by Subarachnoid Hemorrhage in Rats. Neurochem. Res. 2017, 42, 1267–1278. [Google Scholar] [CrossRef] [Green Version]
  107. Hao, G.; Dong, Y.; Huo, R.; Wen, K.; Zhang, Y.; Liang, G. Rutin Inhibits Neuroinflammation and Provides Neuroprotection in an Experimental Rat Model of Subarachnoid Hemorrhage, Possibly Through Suppressing the RAGE-NF-kappaB Inflammatory Signaling Pathway. Neurochem. Res. 2016, 41, 1496–1504. [Google Scholar] [CrossRef]
  108. Guo, Z.; Hu, Q.; Xu, L.; Guo, Z.N.; Ou, Y.; He, Y.; Yin, C.; Sun, X.; Tang, J.; Zhang, J.H. Lipoxin A4 Reduces Inflammation Through Formyl Peptide Receptor 2/p38 MAPK Signaling Pathway in Subarachnoid Hemorrhage Rats. Stroke 2016, 47, 490–497. [Google Scholar] [CrossRef] [Green Version]
  109. Vergouwen, M.D.; Vermeulen, M.; Coert, B.A.; Stroes, E.S.; Roos, Y.B. Microthrombosis after aneurysmal subarachnoid hemorrhage: An additional explanation for delayed cerebral ischemia. J. Cereb. Blood Flow Metab. 2008, 28, 1761–1770. [Google Scholar] [CrossRef]
  110. Sehba, F.A.; Hou, J.; Pluta, R.M.; Zhang, J.H. The importance of early brain injury after subarachnoid hemorrhage. Prog. Neurobiol. 2012, 97, 14–37. [Google Scholar]
  111. McBride, D.W.; Blackburn, S.L.; Peeyush, K.T.; Matsumura, K.; Zhang, J.H. The Role of Thromboinflammation in Delayed Cerebral Ischemia after Subarachnoid Hemorrhage. Front. Neurol. 2017, 8, 555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Dienel, A.; Ammassam Veettil, R.; Hong, S.H.; Matsumura, K.; Kumar, T.P.; Yan, Y.; Blackburn, S.L.; Ballester, L.Y.; Marrelli, S.P.; McCullough, L.D.; et al. Microthrombi Correlates With Infarction and Delayed Neurological Deficits After Subarachnoid Hemorrhage in Mice. Stroke 2020, 51, 2249–2254. [Google Scholar] [CrossRef] [PubMed]
  113. Frontera, J.A.; Aledort, L.; Gordon, E.; Egorova, N.; Moyle, H.; Patel, A.; Bederson, J.B.; Sehba, F. Early platelet activation, inflammation and acute brain injury after a subarachnoid hemorrhage: A pilot study. J. Thromb. Haemost. 2012, 10, 711–713. [Google Scholar] [CrossRef]
  114. Friedrich, B.; Muller, F.; Feiler, S.; Scholler, K.; Plesnila, N. Experimental subarachnoid hemorrhage causes early and long-lasting microarterial constriction and microthrombosis: An in-vivo microscopy study. J. Cereb. Blood Flow Metab. 2012, 32, 447–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Siler, D.A.; Gonzalez, J.A.; Wang, R.K.; Cetas, J.S.; Alkayed, N.J. Intracisternal administration of tissue plasminogen activator improves cerebrospinal fluid flow and cortical perfusion after subarachnoid hemorrhage in mice. Transl. Stroke Res. 2014, 5, 227–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sabri, M.; Ai, J.; Lakovic, K.; D’Abbondanza, J.; Ilodigwe, D.; Macdonald, R.L. Mechanisms of microthrombi formation after experimental subarachnoid hemorrhage. Neuroscience 2012, 224, 26–37. [Google Scholar] [CrossRef]
  117. Andereggen, L.; Neuschmelting, V.; von Gunten, M.; Widmer, H.R.; Fandino, J.; Marbacher, S. The role of microclot formation in an acute subarachnoid hemorrhage model in the rabbit. Biomed. Res. Int. 2014, 2014, 161702. [Google Scholar] [CrossRef]
  118. Stein, S.C.; Browne, K.D.; Chen, X.H.; Smith, D.H.; Graham, D.I. Thromboembolism and delayed cerebral ischemia after subarachnoid hemorrhage: An autopsy study. Neurosurgery 2006, 59, 781–787, discussion 787–8. [Google Scholar] [CrossRef]
  119. Boluijt, J.; Meijers, J.C.; Rinkel, G.J.; Vergouwen, M.D. Hemostasis and fibrinolysis in delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage: A systematic review. J. Cereb. Blood Flow Metab. 2015, 35, 724–733. [Google Scholar] [CrossRef]
  120. Chauhan, A.K.; Kisucka, J.; Brill, A.; Walsh, M.T.; Scheiflinger, F.; Wagner, D.D. ADAMTS13: A new link between thrombosis and inflammation. J. Exp. Med. 2008, 205, 2065–2074. [Google Scholar] [CrossRef]
  121. Stoll, G.; Nieswandt, B. Thrombo-inflammation in acute ischaemic stroke-implications for treatment. Nat. Rev. Neurol. 2019, 15, 473–481. [Google Scholar] [CrossRef] [PubMed]
  122. Muroi, C.; Fujioka, M.; Mishima, K.; Irie, K.; Fujimura, Y.; Nakano, T.; Fandino, J.; Keller, E.; Iwasaki, K.; Fujiwara, M. Effect of ADAMTS-13 on cerebrovascular microthrombosis and neuronal injury after experimental subarachnoid hemorrhage. J. Thromb. Haemost. 2014, 12, 505–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Vergouwen, M.D.; Knaup, V.L.; Roelofs, J.J.; de Boer, O.J.; Meijers, J.C. Effect of recombinant ADAMTS-13 on microthrombosis and brain injury after experimental subarachnoid hemorrhage. J. Thromb. Haemost. 2014, 12, 943–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wan, H.; Wang, Y.; Ai, J.; Brathwaite, S.; Ni, H.; Macdonald, R.L.; Hol, E.M.; Meijers, J.C.M.; Vergouwen, M.D.I. Role of von Willebrand factor and ADAMTS-13 in early brain injury after experimental subarachnoid hemorrhage. J. Thromb. Haemost. 2018, 16, 1413–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Vergouwen, M.D.; Bakhtiari, K.; van Geloven, N.; Vermeulen, M.; Roos, Y.B.; Meijers, J.C. Reduced ADAMTS13 activity in delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. J. Cereb. Blood Flow Metab. 2009, 29, 1734–1741. [Google Scholar] [CrossRef] [PubMed]
  126. Putzer, A.S.; Worthmann, H.; Grosse, G.M.; Goetz, F.; Martens-Lobenhoffer, J.; Dirks, M.; Kielstein, J.T.; Lichtinghagen, R.; Budde, U.; Bode-Boger, S.M.; et al. ADAMTS13 activity is associated with early neurological improvement in acute ischemic stroke patients treated with intravenous thrombolysis. J. Thromb. Thrombolysis 2020, 49, 67–74. [Google Scholar] [CrossRef] [PubMed]
  127. Chou, S.H.; Macdonald, R.L.; Keller, E.; Unruptured Intracranial, A.; Investigators, S.C.P. Biospecimens and Molecular and Cellular Biomarkers in Aneurysmal Subarachnoid Hemorrhage Studies: Common Data Elements and Standard Reporting Recommendations. Neurocrit Care 2019, 30 (Suppl. 1), 46–59. [Google Scholar] [CrossRef]
  128. Jabbarli, R.; Pierscianek, D.; Darkwah Oppong, M.; Sato, T.; Dammann, P.; Wrede, K.H.; Kaier, K.; Kohrmann, M.; Forsting, M.; Kleinschnitz, C.; et al. Laboratory biomarkers of delayed cerebral ischemia after subarachnoid hemorrhage: A systematic review. Neurosurg Rev. 2020, 43, 825–833. [Google Scholar] [CrossRef]
  129. Albert-Weissenberger, C.; Siren, A.L.; Kleinschnitz, C. Ischemic stroke and traumatic brain injury: The role of the kallikrein-kinin system. Prog. Neurobiol. 2013, 101–102, 65–82. [Google Scholar] [CrossRef]
  130. De Meyer, S.F.; Denorme, F.; Langhauser, F.; Geuss, E.; Fluri, F.; Kleinschnitz, C. Thromboinflammation in Stroke Brain Damage. Stroke 2016, 47, 1165–1172. [Google Scholar] [CrossRef] [Green Version]
  131. Kleinschnitz, C.; Stoll, G.; Bendszus, M.; Schuh, K.; Pauer, H.U.; Burfeind, P.; Renne, C.; Gailani, D.; Nieswandt, B.; Renne, T. Targeting coagulation factor XII provides protection from pathological thrombosis in cerebral ischemia without interfering with hemostasis. J. Exp. Med. 2006, 203, 513–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Hagedorn, I.; Schmidbauer, S.; Pleines, I.; Kleinschnitz, C.; Kronthaler, U.; Stoll, G.; Dickneite, G.; Nieswandt, B. Factor XIIa inhibitor recombinant human albumin Infestin-4 abolishes occlusive arterial thrombus formation without affecting bleeding. Circulation 2010, 121, 1510–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Heydenreich, N.; Nolte, M.W.; Gob, E.; Langhauser, F.; Hofmeister, M.; Kraft, P.; Albert-Weissenberger, C.; Brede, M.; Varallyay, C.; Gobel, K.; et al. C1-inhibitor protects from brain ischemia-reperfusion injury by combined antiinflammatory and antithrombotic mechanisms. Stroke 2012, 43, 2457–2467. [Google Scholar] [CrossRef] [Green Version]
  134. Albert-Weissenberger, C.; Mencl, S.; Schuhmann, M.K.; Salur, I.; Gob, E.; Langhauser, F.; Hopp, S.; Hennig, N.; Meuth, S.G.; Nolte, M.W.; et al. C1-Inhibitor protects from focal brain trauma in a cortical cryolesion mice model by reducing thrombo-inflammation. Front. Cell Neurosci. 2014, 8, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Hopp, S.; Albert-Weissenberger, C.; Mencl, S.; Bieber, M.; Schuhmann, M.K.; Stetter, C.; Nieswandt, B.; Schmidt, P.M.; Monoranu, C.M.; Alafuzoff, I.; et al. Targeting coagulation factor XII as a novel therapeutic option in brain trauma. Ann. Neurol. 2016, 79, 970–982. [Google Scholar] [CrossRef] [Green Version]
  136. Hopp, S.; Nolte, M.W.; Stetter, C.; Kleinschnitz, C.; Siren, A.L.; Albert-Weissenberger, C. Alleviation of secondary brain injury, posttraumatic inflammation, and brain edema formation by inhibition of factor XIIa. J. Neuroinflamm. 2017, 14, 39. [Google Scholar] [CrossRef] [Green Version]
  137. Gobel, K.; Pankratz, S.; Asaridou, C.M.; Herrmann, A.M.; Bittner, S.; Merker, M.; Ruck, T.; Glumm, S.; Langhauser, F.; Kraft, P.; et al. Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells. Nat. Commun. 2016, 7, 11626. [Google Scholar] [CrossRef] [Green Version]
  138. Revenko, A.S.; Gao, D.; Crosby, J.R.; Bhattacharjee, G.; Zhao, C.; May, C.; Gailani, D.; Monia, B.P.; MacLeod, A.R. Selective depletion of plasma prekallikrein or coagulation factor XII inhibits thrombosis in mice without increased risk of bleeding. Blood 2011, 118, 5302–5311. [Google Scholar] [CrossRef] [Green Version]
  139. Simao, F.; Ustunkaya, T.; Clermont, A.C.; Feener, E.P. Plasma kallikrein mediates brain hemorrhage and edema caused by tissue plasminogen activator therapy in mice after stroke. Blood 2017, 129, 2280–2290. [Google Scholar] [CrossRef] [PubMed]
  140. Albert-Weissenberger, C.; Hopp, S.; Nieswandt, B.; Siren, A.L.; Kleinschnitz, C.; Stetter, C. How is the formation of microthrombi after traumatic brain injury linked to inflammation? J. Neuroimmunol. 2019, 326, 9–13. [Google Scholar] [CrossRef]
  141. Kleinschnitz, C.; Pozgajova, M.; Pham, M.; Bendszus, M.; Nieswandt, B.; Stoll, G. Targeting platelets in acute experimental stroke: Impact of glycoprotein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and intracranial bleeding. Circulation 2007, 115, 2323–2330. [Google Scholar] [CrossRef] [Green Version]
  142. Kraft, P.; Schuhmann, M.K.; Fluri, F.; Lorenz, K.; Zernecke, A.; Stoll, G.; Nieswandt, B.; Kleinschnitz, C. Efficacy and Safety of Platelet Glycoprotein Receptor Blockade in Aged and Comorbid Mice With Acute Experimental Stroke. Stroke 2015, 46, 3502–3506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Schuhmann, M.K.; Guthmann, J.; Stoll, G.; Nieswandt, B.; Kraft, P.; Kleinschnitz, C. Blocking of platelet glycoprotein receptor Ib reduces “thrombo-inflammation” in mice with acute ischemic stroke. J. Neuroinflamm. 2017, 14, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Schuhmann, M.K.; Stoll, G.; Bieber, M.; Vogtle, T.; Hofmann, S.; Klaus, V.; Kraft, P.; Seyhan, M.; Kollikowski, A.M.; Papp, L.; et al. CD84 Links T Cell and Platelet Activity in Cerebral Thrombo-Inflammation in Acute Stroke. Circ. Res. 2020, 127, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  145. Suzuki, S.; Iwabuchi, T.; Tanaka, T.; Kanayama, S.; Ottomo, M.; Hatanaka, M.; Aihara, H. Prevention of cerebral vasospasm with OKY-046 an imidazole derivative and a thromboxane synthetase inhibitor. A preliminary co-operative clinical study. Acta Neurochir. (Wien.) 1985, 77, 133–141. [Google Scholar] [CrossRef]
  146. Tokiyoshi, K.; Ohnishi, T.; Nii, Y. Efficacy and toxicity of thromboxane synthetase inhibitor for cerebral vasospasm after subarachnoid hemorrhage. Surg. Neurol. 1991, 36, 112–118. [Google Scholar] [CrossRef]
  147. Hirashima, Y.; Endo, S.; Kato, R.; Takaku, A. Prevention of cerebrovasospasm following subarachnoid hemorrhage in rabbits by the platelet-activating factor antagonist, E5880. J. Neurosurg. 1996, 84, 826–830. [Google Scholar] [CrossRef] [Green Version]
  148. Hirashima, Y.; Endo, S.; Nukui, H.; Kobayashi, N.; Takaku, A. Effect of a platelet-activating factor receptor antagonist, E5880, on cerebral vasospasm after aneurysmal subarachnoid hemorrhage--open clinical trial to investigate efficacy and safety. Neurol. Med. Chir. (Tokyo) 2001, 41, 165–175, discussion 175–6. [Google Scholar] [CrossRef] [Green Version]
  149. Lagier, D.; Tonon, D.; Garrigue, P.; Guillet, B.; Giacomino, L.; Martin, J.C.; Alessi, M.C.; Bruder, N.; Velly, L.J. Thromboxane-prostaglandin receptor antagonist, terutroban, prevents neurovascular events after subarachnoid haemorrhage: A nanoSPECT study in rats. Crit Care 2019, 23, 42. [Google Scholar] [CrossRef]
  150. Shi, L.; Xu, L.; Shi, L.; Brandon, D.; Chen, S.; Zhang, J. Intraventricular Recombinant Tissue Plasminogen Activator in Treatment of Aneurysmal Intraventricular Hemorrhage: A Meta-Analysis. Curr. Drug Targets 2017, 18, 1399–1407. [Google Scholar] [CrossRef]
  151. Vergouwen, M.D.; Meijers, J.C.; Geskus, R.B.; Coert, B.A.; Horn, J.; Stroes, E.S.; van der Poll, T.; Vermeulen, M.; Roos, Y.B. Biologic effects of simvastatin in patients with aneurysmal subarachnoid hemorrhage: A double-blind, placebo-controlled randomized trial. J. Cereb. Blood Flow Metab. 2009, 29, 1444–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Zhao, J.; Zhou, D.; Guo, J.; Ren, Z.; Zhou, L.; Wang, S.; Zhang, Y.; Xu, B.; Zhao, K.; Wang, R.; et al. Fasudil Aneurysmal Subarachnoid Hemorrhage Study, G. Efficacy and safety of fasudil in patients with subarachnoid hemorrhage: Final results of a randomized trial of fasudil versus nimodipine. Neurol. Med. Chir. (Tokyo) 2011, 51, 679–683. [Google Scholar] [CrossRef] [Green Version]
  153. James, R.F.; Kramer, D.R.; Aljuboori, Z.S.; Parikh, G.; Adams, S.W.; Eaton, J.C.; Abou Al-Shaar, H.; Badjatia, N.; Mack, W.J.; Simard, J.M. Novel Treatments in Neuroprotection for Aneurysmal Subarachnoid Hemorrhage. Curr. Treat. Options Neurol. 2016, 18, 38. [Google Scholar] [CrossRef]
  154. Li, L.; Lou, X.; Zhang, K.; Yu, F.; Zhao, Y.; Jiang, P. Hydrochloride fasudil attenuates brain injury in ICH rats. Transl. Neurosci. 2020, 11, 75–86. [Google Scholar] [CrossRef]
  155. Vagnozzi, R.; Signoretti, S.; Cristofori, L.; Alessandrini, F.; Floris, R.; Isgro, E.; Ria, A.; Marziali, S.; Zoccatelli, G.; Tavazzi, B.; et al. Assessment of metabolic brain damage and recovery following mild traumatic brain injury: A multicentre, proton magnetic resonance spectroscopic study in concussed patients. Brain 2010, 133, 3232–3242. [Google Scholar] [CrossRef]
  156. Bederson, J.B.; Germano, I.M.; Guarino, L. Cortical blood flow and cerebral perfusion pressure in a new noncraniotomy model of subarachnoid hemorrhage in the rat. Stroke 1995, 26, 1086–1091. [Google Scholar] [CrossRef]
  157. Oddo, M.; Levine, J.M.; Frangos, S.; Maloney-Wilensky, E.; Carrera, E.; Daniel, R.T.; Levivier, M.; Magistretti, P.J.; LeRoux, P.D. Brain lactate metabolism in humans with subarachnoid hemorrhage. Stroke 2012, 43, 1418–1421. [Google Scholar] [CrossRef] [Green Version]
  158. Sehba, F.A.; Bederson, J.B. Mechanisms of acute brain injury after subarachnoid hemorrhage. Neurol. Res. 2006, 28, 381–398. [Google Scholar] [CrossRef]
  159. Sarrafzadeh, A.S.; Sakowitz, O.W.; Lanksch, W.R.; Unterberg, A.W. Time course of various interstitial metabolites following subarachnoid hemorrhage studied by on-line microdialysis. Acta Neurochir. Suppl. 2001, 77, 145–147. [Google Scholar]
  160. Cesarini, K.G.; Enblad, P.; Ronne-Engstrom, E.; Marklund, N.; Salci, K.; Nilsson, P.; Hardemark, H.G.; Hillered, L.; Persson, L. Early cerebral hyperglycolysis after subarachnoid haemorrhage correlates with favourable outcome. Acta Neurochir. 2002, 144, 1121–1131. [Google Scholar] [CrossRef] [PubMed]
  161. Sarrafzadeh, A.; Haux, D.; Plotkin, M.; Ludemann, L.; Amthauer, H.; Unterberg, A. Bedside microdialysis reflects dysfunction of cerebral energy metabolism in patients with aneurysmal subarachnoid hemorrhage as confirmed by 15 O-H2 O-PET and 18 F-FDG-PET. J. Neuroradiol. 2005, 32, 348–351. [Google Scholar] [CrossRef]
  162. Carpenter, D.A.; Grubb, R.L., Jr.; Tempel, L.W.; Powers, W.J. Cerebral oxygen metabolism after aneurysmal subarachnoid hemorrhage. J. Cereb. Blood Flow Metab. 1991, 11, 837–844. [Google Scholar] [CrossRef] [Green Version]
  163. Westermaier, T.; Jauss, A.; Eriskat, J.; Kunze, E.; Roosen, K. Time-course of cerebral perfusion and tissue oxygenation in the first 6 h after experimental subarachnoid hemorrhage in rats. J. Cereb. Blood Flow Metab. 2009, 29, 771–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Liu, F.; Lu, J.; Manaenko, A.; Tang, J.; Hu, Q. Mitochondria in Ischemic Stroke: New Insight and Implications. Aging Dis. 2018, 9, 924–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Hayakawa, K.; Esposito, E.; Wang, X.; Terasaki, Y.; Liu, Y.; Xing, C.; Ji, X.; Lo, E.H. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 2016, 535, 551–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Chou, S.H.; Lan, J.; Esposito, E.; Ning, M.; Balaj, L.; Ji, X.; Lo, E.H.; Hayakawa, K. Extracellular Mitochondria in Cerebrospinal Fluid and Neurological Recovery After Subarachnoid Hemorrhage. Stroke 2017, 48, 2231–2237. [Google Scholar] [CrossRef] [PubMed]
  167. Youn, D.H.; Kim, B.J.; Kim, Y.; Jeon, J.P. Extracellular Mitochondrial Dysfunction in Cerebrospinal Fluid of Patients with Delayed Cerebral Ischemia after Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care 2020, 33, 422–428. [Google Scholar] [CrossRef]
  168. Macdonald, R.L. Origins of the Concept of Vasospasm. Stroke 2016, 47, e11–e15. [Google Scholar] [CrossRef] [Green Version]
  169. Etminan, N.; Vergouwen, M.D.; Ilodigwe, D.; Macdonald, R.L. Effect of pharmaceutical treatment on vasospasm, delayed cerebral ischemia, and clinical outcome in patients with aneurysmal subarachnoid hemorrhage: A systematic review and meta-analysis. J. Cereb. Blood Flow Metab. 2011, 31, 1443–1451. [Google Scholar] [CrossRef] [Green Version]
  170. Rowland, M.J.; Hadjipavlou, G.; Kelly, M.; Westbrook, J.; Pattinson, K.T. Delayed cerebral ischaemia after subarachnoid haemorrhage: Looking beyond vasospasm. Br. J. Anaesth 2012, 109, 315–329. [Google Scholar] [CrossRef] [Green Version]
  171. Geraghty, J.R.; Testai, F.D. Delayed Cerebral Ischemia after Subarachnoid Hemorrhage: Beyond Vasospasm and Towards a Multifactorial Pathophysiology. Curr. Atheroscler. Rep. 2017, 19, 50. [Google Scholar] [CrossRef]
  172. Helbok, R.; Kofler, M.; Schiefecker, A.J.; Gaasch, M.; Rass, V.; Pfausler, B.; Beer, R.; Schmutzhard, E. Clinical Use of Cerebral Microdialysis in Patients with Aneurysmal Subarachnoid Hemorrhage-State of the Art. Front. Neurol. 2017, 8, 565. [Google Scholar] [CrossRef] [Green Version]
  173. Samuelsson, C.; Hillered, L.; Enblad, P.; Ronne-Engstrom, E. Microdialysis patterns in subarachnoid hemorrhage patients with focus on ischemic events and brain interstitial glutamine levels. Acta Neurochir (Wien.) 2009, 151, 437–446, discussion 446. [Google Scholar] [CrossRef]
  174. Helbok, R.; Madineni, R.C.; Schmidt, M.J.; Kurtz, P.; Fernandez, L.; Ko, S.B.; Choi, A.; Stuart, M.R.; Connolly, E.S.; Lee, K.; et al. Intracerebral monitoring of silent infarcts after subarachnoid hemorrhage. Neurocrit. Care 2011, 14, 162–167. [Google Scholar] [CrossRef]
  175. Patet, C.; Quintard, H.; Zerlauth, J.B.; Maibach, T.; Carteron, L.; Suys, T.; Bouzat, P.; Bervini, D.; Levivier, M.; Daniel, R.T.; et al. Bedside cerebral microdialysis monitoring of delayed cerebral hypoperfusion in comatose patients with poor grade aneurysmal subarachnoid haemorrhage. J. Neurol. Neurosurg. Psychiatry 2017, 88, 332–338. [Google Scholar] [CrossRef]
  176. Dorhout Mees, S.M.; Rinkel, G.J.; Feigin, V.L.; Algra, A.; van den Bergh, W.M.; Vermeulen, M.; van Gijn, J. Calcium antagonists for aneurysmal subarachnoid haemorrhage. Cochrane Database Syst Rev. 2007, 2007, CD000277. [Google Scholar] [CrossRef]
  177. Stacpoole, P.W.; Henderson, G.N.; Yan, Z.; James, M.O. Clinical pharmacology and toxicology of dichloroacetate. Env. Health Perspect 1998, 106 (Suppl. 4), 989–994. [Google Scholar]
  178. Thibodeau, A.; Geng, X.; Previch, L.E.; Ding, Y. Pyruvate dehydrogenase complex in cerebral ischemia-reperfusion injury. Brain Circ. 2016, 2, 61–66. [Google Scholar]
  179. Wang, P.; Chen, M.; Yang, Z.; Yu, T.; Zhu, J.; Zhou, L.; Lin, J.; Fang, X.; Huang, Z.; Jiang, L.; et al. Activation of Pyruvate Dehydrogenase Activity by Dichloroacetate Improves Survival and Neurologic Outcomes After Cardiac Arrest in Rats. Shock 2018, 49, 704–711. [Google Scholar] [CrossRef]
  180. Kho, A.R.; Choi, B.Y.; Lee, S.H.; Hong, D.K.; Jeong, J.H.; Kang, B.S.; Kang, D.H.; Park, K.H.; Park, J.B.; Suh, S.W. The Effects of Sodium Dichloroacetate on Mitochondrial Dysfunction and Neuronal Death Following Hypoglycemia-Induced Injury. Cells 2019, 8, 405. [Google Scholar] [CrossRef] [Green Version]
  181. Jalal, F.Y.; Bohlke, M.; Maher, T.J. Acetyl-L-carnitine reduces the infarct size and striatal glutamate outflow following focal cerebral ischemia in rats. Ann. N. Y. Acad. Sci. 2010, 1199, 95–104. [Google Scholar] [CrossRef]
  182. Martin, E.; Rosenthal, R.E.; Fiskum, G. Pyruvate dehydrogenase complex: Metabolic link to ischemic brain injury and target of oxidative stress. J. Neurosci. Res. 2005, 79, 240–247. [Google Scholar] [CrossRef] [Green Version]
  183. Rosenthal, R.E.; Bogaert, Y.E.; Fiskum, G. Delayed therapy of experimental global cerebral ischemia with acetyl-L-carnitine in dogs. Neurosci. Lett. 2005, 378, 82–87. [Google Scholar] [CrossRef]
  184. Zanelli, S.A.; Solenski, N.J.; Rosenthal, R.E.; Fiskum, G. Mechanisms of ischemic neuroprotection by acetyl-L-carnitine. Ann. N. Y. Acad. Sci. 2005, 1053, 153–161. [Google Scholar] [CrossRef] [Green Version]
  185. Rosenthal, R.E.; Williams, R.; Bogaert, Y.E.; Getson, P.R.; Fiskum, G. Prevention of postischemic canine neurological injury through potentiation of brain energy metabolism by acetyl-L-carnitine. Stroke 1992, 23, 1312–1317. [Google Scholar] [CrossRef] [Green Version]
  186. Wu, H.; Niu, H.; Wu, C.; Li, Y.; Wang, K.; Zhang, J.; Wang, Y.; Yang, S. The autophagy-lysosomal system in subarachnoid haemorrhage. J. Cell Mol. Med. 2016, 20, 1770–1778. [Google Scholar] [CrossRef]
  187. Chen, Y.; Huang, L.; Zhang, H.; Diao, X.; Zhao, S.; Zhou, W. Reduction in Autophagy by (-)-Epigallocatechin-3-Gallate (EGCG): A Potential Mechanism of Prevention of Mitochondrial Dysfunction After Subarachnoid Hemorrhage. Mol. Neurobiol. 2017, 54, 392–405. [Google Scholar] [CrossRef]
  188. Daou, B.J.; Koduri, S.; Thompson, B.G.; Chaudhary, N.; Pandey, A.S. Clinical and experimental aspects of aneurysmal subarachnoid hemorrhage. Cns Neurosci. 2019, 25, 1096–1112. [Google Scholar] [CrossRef]
  189. Van Lieshout, J.H.; Dibué-Adjei, M.; Cornelius, J.F.; Slotty, P.J.; Schneider, T.; Restin, T.; Boogaarts, H.D.; Steiger, H.J.; Petridis, A.K.; Kamp, M.A. An introduction to the pathophysiology of aneurysmal subarachnoid hemorrhage. Neurosurg Rev. 2018, 41, 917–930. [Google Scholar] [CrossRef]
  190. Vatter, H.; Zimmermann, M.; Tesanovic, V.; Raabe, A.; Schilling, L.; Seifert, V. Cerebrovascular characterization of clazosentan, the first nonpeptide endothelin receptor antagonist clinically effective for the treatment of cerebral vasospasm. Part I: Inhibitory effect on endothelin(A) receptor-mediated contraction. J. Neurosurg. 2005, 102, 1101–1107. [Google Scholar] [CrossRef]
  191. Macdonald, R.L.; Kassell, N.F.; Mayer, S.; Ruefenacht, D.; Schmiedek, P.; Weidauer, S.; Frey, A.; Roux, S.; Pasqualin, A.; CONSCIOUS-1 Investigators. Clazosentan to overcome neurological ischemia and infarction occurring after subarachnoid hemorrhage (CONSCIOUS-1): Randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke 2008, 39, 3015–3021. [Google Scholar] [CrossRef] [Green Version]
  192. Macdonald, R.L.; Higashida, R.T.; Keller, E.; Mayer, S.A.; Molyneux, A.; Raabe, A.; Vajkoczy, P.; Wanke, I.; Bach, D.; Frey, A.; et al. Clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid haemorrhage undergoing surgical clipping: A randomised, double-blind, placebo-controlled phase 3 trial (CONSCIOUS-2). Lancet Neurol. 2011, 10, 618–625. [Google Scholar] [CrossRef]
  193. Macdonald, R.L.; Higashida, R.T.; Keller, E.; Mayer, S.A.; Molyneux, A.; Raabe, A.; Vajkoczy, P.; Wanke, I.; Bach, D.; Frey, A.; et al. Randomized trial of clazosentan in patients with aneurysmal subarachnoid hemorrhage undergoing endovascular coiling. Stroke 2012, 43, 1463–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. van den Bergh, W.M.; Algra, A.; van der Sprenkel, J.W.; Tulleken, C.A.; Rinkel, G.J. Hypomagnesemia after aneurysmal subarachnoid hemorrhage. Neurosurgery 2003, 52, 276–281, discussion 281–2. [Google Scholar] [CrossRef] [PubMed]
  195. Westermaier, T.; Stetter, C.; Kunze, E.; Willner, N.; Raslan, F.; Vince, G.H.; Ernestus, R.I. Magnesium treatment for neuroprotection in ischemic diseases of the brain. Exp. Transl. Stroke Med. 2013, 5, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. van den Bergh, W.M.; Algra, A.; van Kooten, F.; Dirven, C.M.; van Gijn, J.; Vermeulen, M.; Rinkel, G.J. Magnesium sulfate in aneurysmal subarachnoid hemorrhage: A randomized controlled trial. Stroke 2005, 36, 1011–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Stippler, M.; Crago, E.; Levy, E.I.; Kerr, M.E.; Yonas, H.; Horowitz, M.B.; Kassam, A. Magnesium infusion for vasospasm prophylaxis after subarachnoid hemorrhage. J. Neurosurg. 2006, 105, 723–729. [Google Scholar] [CrossRef] [Green Version]
  198. Kunze, E.; Lilla, N.; Stetter, C.; Ernestus, R.I.; Westermaier, T. Magnesium Protects in Episodes of Critical Perfusion after Aneurysmal SAH. Transl. Neurosci. 2018, 9, 99–105. [Google Scholar] [CrossRef]
  199. Westermaier, T.; Stetter, C.; Vince, G.H.; Pham, M.; Tejon, J.P.; Eriskat, J.; Kunze, E.; Matthies, C.; Ernestus, R.I.; Solymosi, L.; et al. Prophylactic intravenous magnesium sulfate for treatment of aneurysmal subarachnoid hemorrhage: A randomized, placebo-controlled, clinical study. Crit Care Med. 2010, 38, 1284–1290. [Google Scholar] [CrossRef]
  200. Dorhout Mees, S.M.; Algra, A.; Vandertop, W.P.; van Kooten, F.; Kuijsten, H.A.; Boiten, J.; van Oostenbrugge, R.J.; Al-Shahi Salman, R.; Lavados, P.M.; Rinkel, G.J.; et al. Magnesium for aneurysmal subarachnoid haemorrhage (MASH-2): A randomised placebo-controlled trial. Lancet 2012, 380, 44–49. [Google Scholar] [CrossRef] [Green Version]
  201. Wong, G.K.; Poon, W.S.; Chan, M.T.; Boet, R.; Gin, T.; Ng, S.C.; Zee, B.C. Intravenous magnesium sulphate for aneurysmal subarachnoid hemorrhage (IMASH): A randomized, double-blinded, placebo-controlled, multicenter phase III trial. Stroke 2010, 41, 921–926. [Google Scholar] [CrossRef]
  202. Pickard, J.D.; Murray, G.D.; Illingworth, R.; Shaw, M.D.; Teasdale, G.M.; Foy, P.M.; Humphrey, P.R.; Lang, D.A.; Nelson, R.; Richards, P.; et al. Effect of oral nimodipine on cerebral infarction and outcome after subarachnoid haemorrhage: British aneurysm nimodipine trial. BMJ 1989, 298, 636–642. [Google Scholar] [CrossRef] [Green Version]
  203. Amory, C.F.; Varelas, P.N. Magnesium and Hydrogen in Subarachnoid Hemorrhage: Is Neuroprotection Finally a Reality? Stroke 2021, 52, 28–30. [Google Scholar] [CrossRef]
  204. Takeuchi, S.; Kumagai, K.; Toyooka, T.; Otani, N.; Wada, K.; Mori, K. Intravenous Hydrogen Therapy With Intracisternal Magnesium Sulfate Infusion in Severe Aneurysmal Subarachnoid Hemorrhage. Stroke 2021, 52, 20–27. [Google Scholar] [CrossRef]
  205. Westermaier, T.; Stetter, C.; Kunze, E.; Willner, N.; Holzmeier, J.; Kilgenstein, C.; Lee, J.Y.; Ernestus, R.I.; Roewer, N.; Muellenbach, R.M. Controlled transient hypercapnia: A novel approach for the treatment of delayed cerebral ischemia after subarachnoid hemorrhage? J. Neurosurg. 2014, 121, 1056–1062. [Google Scholar] [CrossRef] [Green Version]
  206. Westermaier, T.; Stetter, C.; Kunze, E.; Willner, N.; Holzmeier, J.; Weiland, J.; Koehler, S.; Lotz, C.; Kilgenstein, C.; Ernestus, R.I.; et al. Controlled Hypercapnia Enhances Cerebral Blood Flow and Brain Tissue Oxygenation After Aneurysmal Subarachnoid Hemorrhage: Results of a Phase 1 Study. Neurocrit. Care 2016, 25, 205–214. [Google Scholar] [CrossRef] [PubMed]
  207. Petridis, A.K.; Doukas, A.; Kienke, S.; Maslehaty, H.; Mahvash, M.; Barth, H.; Mehdorn, H.M. The effect of lung-protective permissive hypercapnia in intracerebral pressure in patients with subarachnoid haemorrhage and ARDS. A retrospective study. Acta Neurochir. (Wien.) 2010, 152, 2143–2145. [Google Scholar] [CrossRef]
  208. Lilla, N.; Rinne, C.; Weiland, J.; Linsenmann, T.; Ernestus, R.I.; Westermaier, T. Early Transient Mild Hypothermia Attenuates Neurologic Deficits and Brain Damage After Experimental Subarachnoid Hemorrhage in Rats. World Neurosurg. 2018, 109, e88–e98. [Google Scholar] [CrossRef]
  209. Török, E.; Klopotowski, M.; Trabold, R.; Thal, S.C.; Plesnila, N.; Schöller, K. Mild hypothermia (33 degrees C) reduces intracranial hypertension and improves functional outcome after subarachnoid hemorrhage in rats. Neurosurgery 2009, 65, 352–359. [Google Scholar] [CrossRef]
  210. Ma, J.; Wang, Z.; Liu, C.; Shen, H.; Chen, Z.; Yin, J.; Zuo, G.; Duan, X.; Li, H.; Chen, G. Pramipexole-Induced Hypothermia Reduces Early Brain Injury via PI3K/AKT/GSK3β pathway in Subarachnoid Hemorrhage rats. Sci Rep. 2016, 6, 23817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Seule, M.; Muroi, C.; Sikorski, C.; Hugelshofer, M.; Winkler, K.; Keller, E. Therapeutic hypothermia reduces middle cerebral artery flow velocity in patients with severe aneurysmal subarachnoid hemorrhage. Neurocrit. Care 2014, 20, 255–262. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Summary of neuroprotective targets, potential agents and therapeutic strategies in different compartments following aSAH.
Figure 1. Summary of neuroprotective targets, potential agents and therapeutic strategies in different compartments following aSAH.
Ijms 22 05442 g001
Table 1. Potential neuroprotective therapies targeting neuroinflammation.
Table 1. Potential neuroprotective therapies targeting neuroinflammation.
Therapeutic AgentTargetModelOutcome Measures/Findings Reference
LPS, PLX3397MicrogliaExperimental aSAH in miceReduction in neuronal cell deathHeinz, R. et al., 2021 [29]
Interleukin-2 (IL-2)Regulatory T-cellsExperimental aSAH in ratsReduction in neuronal injury and proinflammatory factors, increase in neuronal functionsDong et al., 2021 [71]
Mesenchymal stem cell-derived extracellular vesiclesMicroglial M2 polarizationExperimental aSAH in ratsReduction in inflammatory cytokines and inflammation, increase in neuroprotective effectsHan et al., 2021 [72]
Adenosine A3 receptor agonistMicroglial polarizationExperimental aSAH in ratsIncrease in anti-inflammatory and neuroprotective effectsLi et al., 2020 [73]
Milk fat globule-epidural growth factor (MFG-EP)Microglial polarizationExperimental aSAH in miceReduction in brain edema and proinflammatory factors, increase in neurological factorsGao et al., 2021 [74]
Mixture of gas containing argonMicroglial inflammatory responseExperimental aSAH in ratsReduction in early hippocampal neuronal damageKremer et al., 2020 [75]
EPZ6438 (specific EZH2 inhibitor)EZH2 (enhancer of zeste homolog 2)Experimental aSAH in ratsReduction in attenuated neuroinflammatory brain injuryLuo et al., 2020 [76]
Oxyhemoglobin (OxyHb)RNF26 (regulating TLRs)Experimental aSAH in ratsSilence: reduction in neuronal injury and neurological dysfunctionChen et al., 2020 [77]
LP-17TREM-1 myeloid cellsExperimental aSAH in miceAmelioration of microglial pyroptosisXu et al., 2020 [78]
Resolvin D1Lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) in microgliaExperimental aSAH in ratsInhibiting H6 promoted microglial pro-inflammatory polarization, neuronal oxidant damage and deathLiu et al., 2020 [79]
Hydrogen sulfide (H2S)TLR4/NF-κB pathway in microglia Experimental aSAH in ratsReduction in cognitive impairment and amelioration of neuroinflammation in microgliaDuan et al., 2020 [80]
Oxyhemoglobin (OxyHb)CC chemokine ligand 20 (CCL20)Experimental aSAH in miceReduction in apoptotic neuronsLiao et al., 2020 [81]
Translocator protein (TSPO) and TSPO ligand Ro5–4864Microglia/macrophages polarizationExperimental aSAH in miceImprovement of neurological function, increase in expression of anti-inflammatory factorsZhou et al., 2020 [82]
Oxyhemoglobin (OxyHb)CCM3 overexpression and NF-κB signaling pathwayExperimental aSAH in ratsReduction in cellular degeneration, neurocognitive impairment and inflammatory factors (TNF-a and IL-1β)Peng et al., 2020 [83]
Modified exosomes (miR-193b-3p)HDAC3, NF-κB1. aSAH patients and healthy controls to define profile
2. experimental aSAH in mice
Reduction in homological behavioral impairment, brain edema and BBB injuryLai et al., 2020 [84]
CurcuminM2 polarization through TLR4/MyD88/NF-κB signaling pathwayExperimental aSAH in tlr4−/− mice and wild type (WT)Alleviation of neuroinflammation response, microglia phenotype shift and release of proinflammatory mediatorsGao et al., 2019 [85]
Dehydroepiandrosterone (DHEA)Microglial activationExperimental aSAH in C57BL/6 miceIncrease in neuroprotective effects, suppression of inflammationTao et al., 2019 [86]
Apelin-13Apelin receptor (APJ)/endoplasmic reticulum stress associated inflammationExperimental aSAH in ratsReduction in oxidative stress and neuroinflammationXu et al., 2019 [87]
BMS-470539Melanocortin 1 receptor (MC1R)Experimental aSAH in ratsSuppression of microglial activation and neutrophil infiltrationXu et al., 2020 [88]
TAK 242 (TLR4 antagonist)Toll-like 4 receptor (TLR4)Experimental aSAH in miceSuppression of brain edemaOkada et al., 2020 [89]
BexaroteneRetinoid X receptorExperimental aSAH in ratsDecrease in neuroinflammation, improvement of neurological deficitsZuo et al., 2019 [90]
RP 001 hydrochlorideS1P/S1PR pathwayExperimental aSAH in miceDecrease in neuroinflammation, alleviation of neurological damageLi et al., 2019 [91]
Bone marrow mesenchymal stem cellsNotch 1 signaling pathwayExperimental aSAH in ratsAmelioration of neurobehavioral impairments and BBB disruptionLiu et al., 2019 [92]
FluoxetineTLR4/MYD88/NF-κB pathwayExperimental aSAH in ratsDecrease in BBB disruption and brain edema, improvement of neurological functionLiu et al., 2018 [93]
Apolipoprotein EJak2/STAT3 signaling pathwayExperimental aSAH in miceDecrease in oxidative stress and inflammationPang et al., 2018 [94]
TSG-6Microglial phenotype shift/SOCS3/STAT3 pathwayExperimental aSAH in ratsAmelioration of brain injury, decrease in proinflammatory mediatorsLi et al., 2018 [95]
TAT-Pep5PResident microglia, p75 neurotrophin receptor (p75NTR)Experimental aSAH in transgenic miceReduction in microglial activation, neuroinflammation and EBIXu et al., 2019 [96]
MST1 inhibitor XMU-MP-1MST1, NF-κB/MMP-9 pathwayExperimental aSAH in miceAlleviation of neurological deficits, BBB, brain edema, neuroinflammation and white matter injuryQu et al., 2018 [97]
rh-Aggf1PI3K/Akt/NF-κB pathwayExperimental aSAH in ratsDecrease in neuroinflammation and BBB disruption, improvement of neurological deficitsZhu et al., 2018 [98]
IAXO-102 (TLR4 antagonist)TLR4Experimental aSAH in C57BL/6 miceReduction in neurological impairments, brain edema, BBB disruption, increase in survival ratesOkada et al., 2019 [99]
BexarotenePPARγExperimental aSAH in C57BL/6 miceIncrease in neurological function, reduction in neuronal cell death and microglial activationTu et al., 2018 [100]
FTY720 (PP2A agonist)Tristetraprolin (TTP), protein phosphatase 2A (PP2A)Experimental aSAH in ratsReduction in apoptosis, neuroinflammation and brain edema, increase in neurological functionYin et al., 2018 [101]
Rolipram (specific phosphodiesterase-4 inhibitor)SIRT1/NF-κB pathwayExperimental aSAH in ratsReduction in brain edema, neurological dysfunction and neuronal cell deathPeng et al., 2018 [102]
Human Netrin-1 (rh-NTN-1)UNC5B (receptor of NTN-1)Experimental aSAH in ratsIncrease in neurobehavioral function, reduction in brain edema and microglia activationXie et al., 2018 [103]
Fluoxetine, AC-YVAD-CMK (caspase-inhibitor)NLRP3 inflammasome, caspase-1Experimental aSAH in ratsIncrease in neurological function, reduction in brain edema and autophagy activationLi et al., 2017 [104]
Methylene blueAkt/GSK-3β/MEF2D pathwayExperimental aSAH in ratsReduction in neurological dysfunction and brain edemaXu et al., 2017 [105]
IL-1 receptor antagonist (IL-1Ra, anakinra)Interleukin-1 (IL-1)Randomized, open-label, clinical study in aSAH-patientsDifference in plasma IL-6, plasma pharmacokinetics for IL-1Ra, clinical outcome at 6 monthsGalea et al., 2018 [63]
AE1–329 (EP4 selective agonist)Prostanoid 4 receptor (EP4)Experimental aSAH in ratsReduction in neurological dysfunction, BBB damage, brain edema, reactivation of microglia, proinflammatory cytokinesXu et al., 2017 [106]
RutinRAGE- NF-κB inflammatory signaling pathwayExperimental aSAH in ratsIncrease in neurological function, reduction in BBB permeability, brain water content and neuronal cell deathHao et al., 2016 [107]
Exogenous LXA4 (lipoxin A4)Formyl peptide receptor 2 (FPR2), p38 MAPKExperimental aSAH in ratsIncrease in neurological functions, reduction in neutrophil infiltration and brain water contentGuo et al., 2016 [108]
Table 2. Potential neuroprotective pharmacotherapies targeting thromboinflammation.
Table 2. Potential neuroprotective pharmacotherapies targeting thromboinflammation.
Therapeutic AgentTargetModelReference
Thromboxane antagonists, COX1-inhibitors, PAF antagonistsPlatelet aggregationExperimental and clinical aSAHLagier et al. [149], Suzuki et al. [145], Tokiyoshi et al. [146], Hirashima et al. [147,148]
intraventricular thrombolysis (rh tPA)Clot clearanceExperimental and clinical aSAHShi et al. [150]
rh-ADAMTS13vWF-induced thrombosis and inflammationExperimental and clinical aSAHMuroi et al. [122], Vergouwen et al. [123,125], Wan et al. [124], Chauhan et al. [120]
FXIIa inhibitors (C1 inhibitor, rh infestin-4) Contact kinin system (platelet aggregation and neuroinflammation)Experimental stroke and TBIKleinschnitz et al. [131], Hagedorn et al. [132], Heydenreich et al. [133], Hopp et al. [135,136], Albert-Weissenberger et al. [134]
Anti-platelet receptor antibodiesThrombosis, neuroinflammation, immune cellsExperimental stroke and TBIKleinschnitz et al. [141], Schuhmann et al. [143], Albert-Weissenberger et al. [140], Stoll and Nieswandt [121]
Fractionated heparin, glibenclamide, statins, anti-proinflammatory cytokine agentsNeuroinflammationExperimental and clinical aSAHJames et al. [153], McBride et al. [111], Vergouwen et al. [151]
Fasudil (ROCK2 inhibitor)NeuroinflammationExperimental intracerebral hemorrhage (ICH) and clinical aSAHMcBride et al. [111], Li et al. [154], Zhao et al. [152]
NimodipineVasospasms, thrombosis, leukocyte infiltrationExperimental and clinical aSAHMcBride et al. [111]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Weiland, J.; Beez, A.; Westermaier, T.; Kunze, E.; Sirén, A.-L.; Lilla, N. Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int. J. Mol. Sci. 2021, 22, 5442. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115442

AMA Style

Weiland J, Beez A, Westermaier T, Kunze E, Sirén A-L, Lilla N. Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH). International Journal of Molecular Sciences. 2021; 22(11):5442. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115442

Chicago/Turabian Style

Weiland, Judith, Alexandra Beez, Thomas Westermaier, Ekkehard Kunze, Anna-Leena Sirén, and Nadine Lilla. 2021. "Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH)" International Journal of Molecular Sciences 22, no. 11: 5442. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115442

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