Abstract
The loss of cerebral autoregulation (CA) is a common and detrimental secondary injury mechanism following acute brain injury and has been associated with worse morbidity and mortality. However patient outcomes have not as yet been conclusively proven to have improved as a result of CA-directed therapy. While CA monitoring has been used to modify CPP targets, this approach cannot work if the impairment of CA is not simply related to CPP but involves other underlying mechanisms and triggers, which at present are largely unknown. Neuroinflammation, particularly inflammation affecting the cerebral vasculature, is an important cascade that occurs following acute injury. We hypothesise that disturbances to the cerebral vasculature can affect the regulation of CBF, and hence the vascular inflammatory pathways could be a putative mechanism that causes CA dysfunction. This review provides a brief overview of CA, and its impairment following brain injury. We discuss candidate vascular and endothelial markers and what is known about their link to disturbance of the CBF and autoregulation. We focus on human traumatic brain injury (TBI) and subarachnoid haemorrhage (SAH), with supporting evidence from animal work and applicability to wider neurologic diseases.
Keywords: Cerebral autoregulation, cerebral blood flow, biomarkers, brain trauma, inflammation
Introduction
Traumatic brain injury (TBI) and subarachnoid haemorrhage (SAH) are often grouped together as ‘Acute Brain Injuries’ and are responsible for a substantial burden of neurological morbidity and mortality. There are many factors influencing outcome after brain injury and these processes are dynamically changing over the first few days post-injury. Clinical management of patients is focussed on the prevention of secondary brain injury and on providing a physiologically stable environment for brain recovery. One common contribution to secondary brain injury in both TBI and SAH is the loss of cerebral autoregulation (CA).
CA is the brain’s physiological mechanism that stabilises cerebral blood flow (CBF) and ensures that the brain is supplied with adequate nutrients and oxygen despite changes in cerebral perfusion pressure (CPP, the mathematical difference between mean arterial pressure [MAP] – intracranial pressure [ICP]).1,2 In severe brain trauma, fully functional CA guards against derangement in CPP, thereby avoiding ischaemia and hyper-perfusion.3,4 On the other hand, impaired CA has been associated with worsened outcome and mortality in severe TBI5,6 and is understood to be a key factor in the development of secondary injury. Despite developments made in TBI injury dynamic CA monitoring, mortality and outcome seem to have remained relatively unchanged in the last two decades. 7 This may, at least partially, reflect a lack of understanding of the molecular mechanistic underpinnings of CA, and therefore a lack of means of therapeutically modifying CA disturbance. Investigations into the possible mechanisms causing CA impairment is fundamental in directing and developing potential patient therapies and improving overall outcome following injury.
It is widely accepted that neuroinflammation plays a role in brain injury.8–10 In the initial period after injury, the pro-inflammatory response aids in the preservation of surviving neuronal tissue and cell repair.11,12 However, if the inflammatory response is chronic as it is in brain injury, it may be detrimental and harmful to recovery. 13 The neuroinflammatory cascade inflicts direct injury to neurons and supporting cells, but also includes mediators, cytokines and chemokines that set off other potentially detrimental cascades, some or all of which may contribute to secondary brain injury. One such continuous and resultant cascade is vascular inflammation. This may cause and exacerbate vascular pathological states, such as endothelial cell swelling/oedema 14 within cerebral blood vessel walls and increased vascular permeability. These changes result in decreased vascular reactivity and can contribute to the inadequate CBF following brain injury. Neuroinflammation affecting the vasculature provides a promising putative link between the inflammatory pathway and CA disturbances following TBI and SAH.
In this review, we give a brief overview of CA, and what is known regarding its impairment following acute brain injury. We add supporting evidence from animal work where relevant, but mainly focus on human TBI and SAH. Then, we discuss candidate vascular and endothelial markers and what is known about their link to disturbance of the CBF and autoregulation. We acknowledge and recognise the applicability of this work to other neurologic diseases, and briefly discuss the advantage of future work directed at the molecular underpinning of CA dysfunction to multiple fields.
Brief overview of cerebral autoregulation
CA is an important physiological mechanism that ensures adequate CBF despite changes in CPP. If CA is impaired, CBF will fluctuate passively with CPP which puts the already-injured brain at risk for further secondary injury. If CPP, and therefore CBF if CA is impaired, is too low, the brain is at risk of ischaemia; if CPP is too high, there is a risk of aggravated brain oedema resulting in a secondary rise of ICP.
As this is such an important contribution to brain homeostasis, there are several mechanisms by which vascular tone within blood vessels and therefore their calibre is regulated: myogenic, metabolic (including biochemical) and neurogenic. There is significant overlap between these mechanisms, and the contribution of each mechanism is largely unknown.
Genetic variation affecting multiple mechanisms of CBF control
While it is generally accepted that the underlying mechanisms of autoregulation are largely unknown, there are existing postulations and evidence for some mechanisms in the literature. The genetic drivers and polymorphisms that may play an important role in CA following TBI have been discussed in the review by Zeiler et al. 15 This synopsis of investigations of CA mechanisms from a genetic standpoint have highlighted pathways involving inflammation, central autonomic response, and cortical spreading depression. Single-nucleotide polymorphisms (SNPs) relating to endothelial, myogenic, neurotransmitters and metabolic mechanisms in the brain vasculature were identified as contributing to CA. Additionally, the renin-angiotensin system has been involved in modulation of the vascular myogenic responses, with SNPs in genes encoding angiotensin converting enzyme (ACE) and the type 2 angiotensin II receptor (AT2) linking to the development of delayed ischaemic neurological deficits in SAH.16,17 Furthermore, the recessive allele of ACE has been linked to worse outcome scores at 6 months post TBI.15,18 Identifying candidate SNPs may be useful as key pathways are highlighted, but the manifestation of these key genetic pathways in the clinical setting is highly influenced by multiple factors. While genetic investigations are important for identifying fundamental predispositions to CA dysfunction, it is necessary to study CBF control in the clinical setting.
Myogenic
The myogenic mechanism of CA is based on the intrinsic ability of smooth muscle cells in the cerebral blood vessels to constrict and dilate in response to changes in transmural pressure. 19 PRx, the pressure reactivity index, is a mathematical indicator of cerebrovascular pressure reactivity and is calculated as the correlation coefficient between MAP and ICP. PRx is calculated as the moving correlation coefficient between MAP and ICP, and ranges from −1 to 1, with negative values suggesting intact pressure reactivity, and positive values indicating impaired reactivity. 20 Its main assumption is that ICP reflects cerebrovascular volume changes brought about by slow variations in MAP (Figure 1). It has been shown that PRx has a strong and independent association with clinical outcome.4,21
Figure 1.
An overview of cerebral autoregulation. Cerebral autoregulation functions over a range of cerebral perfusion pressures (CPP; the mathematical difference between mean arterial pressure [MAP] and intracranial pressure [ICP]). Blood pressures outside of the autoregulatory zone cause passive changes in cerebral blood flow and puts the patient at risk of secondary injury. One autoregulatory mechanism of cerebral blood flow control is cerebrovascular pressure reactivity. The pressure reactivity index (PRx) is the correlation coefficient between the MAP and ICP, with a negative PRx indicating good pressure reactivity.
The myogenic mechanism of CA is supported by primary observations of cerebral arteries changing diameter in response to blood pressure stimuli. These reactions persist when neurogenic control of the vessels is eliminated.19,22–25 Changes in transmural pressure and blood flow induce diameter changes in cerebral blood vessels, however the exact molecular mechanisms acting on the endothelium are largely unknown.
Calcium signalling, and fluxes in ion concentration is fundamental to the functioning of most cells and plays an important role in the initiation of the myogenic response of CA. Calcium flux in the smooth muscle cells of cerebral vessels lead to eventual vasoconstriction as follows: changes in transmural pressure and wall tension are detected by a mechanoreceptor, which leads to the opening of voltage gated calcium channels. Calcium floods the cell and activates myosin light chain kinase (MLCK). 26 This leads to myosin phosphorylation and actin polymerisation, and smooth muscle cell contraction. Alternatively, rapid dephosphorylation of the myosin heavy chain promotes relaxation. 27
Importantly, while calcium influx to the cell is required for the myogenic response, voltage gated calcium channels is not the only way to increase intracellular calcium. Activation of membrane-bound phospholipase C leads to the production of 1,4,5-triphosphate (IP3) and 1,2 diacylglycerol (DAG). IP3 will then diffuse into the cell and stimulate the sarcoplasmic reticulum to release calcium.27,28
Metabolic
The metabolic mechanism of CA is the mechanism that links the local rate of metabolism to cerebral vasculature, hence controlling CBF. The brain’s energy demand is tightly linked to the blood supply of oxygen and nutrients. 29 Therefore, this mechanism is stimulated due to changes in the local metabolism in the cerebral vasculature, such as cerebral hypercapnia. The brain vasculature is highly sensitive to changes in partial pressure of arterial carbon dioxide (PaCO2). 30 A high PaCO2 will cause vasodilation, resulting in an increase of CBF. This mechanism serves as an automatic feedback loop – increased brain activity and metabolism will lead to an increase in PaCO2, as detected by surrounding neurons. These neurons then produce neuronal nitric oxide synthase (nNOS) which subsequently facilitates vasodilation, and an increase in CBF.31–33 Endothelial-derived NOS (eNOS) also leads to vasodilation (to be discussed later) but the hypercapnia-induced vasodilation seems to be nNOS-dependent.34,35
Importantly, this metabolic mechanism (CO2 reactivity and flow-metabolism coupling) is different from pressure reactivity as it is, largely, CPP-independent. 36 Another important distinction of note is between the metabolic mechanism of CA, and neurovascular coupling. The latter is a mechanism by which CBF is regulated according to local cortical/neuronal activity. 29 The intuitive concept that neuronal substrate demand via increased activity (i.e., increased metabolic demand) is linked to supply (blood flow) is oversimplified, and it is likely that neuronal signalling itself (likely via neurotransmitters and/or extracellular potassium ions) are involved in this mechanism.37,38 There is definite overlap and interaction between these two important CBF regulatory mechanisms, but these terms should not be used interchangeably.
Neurogenic
The neurogenic mechanism of CA refers to the direct control of cerebral vasculature via autonomic innervation. Intrinsic/central innervation of cerebral vessels is likely to have a global effect on CBF control, with modulations from extrinsic innervation (in addition to inflammatory, metabolic, and myogenic influences). Cerebral arteries are extensively innervated, and have been shown to constrict following adrenergic stimulation, and dilate following cholinergic stimulation. However, sympathetic and parasympathetic denervation did not abolish CBF regulation or reactivity to changes in PaCO2.39,40 This implies that the neurogenic mechanism can modulate CBF control, but it is not a primary CA control mechanism. Interestingly, calcium signalling, as in the myogenic response, is also important for neurovascular coupling at the capillary level specifically, as astrocytic calcium is necessary for vasodilation of capillaries.27,41
An important note in the investigations of CBF regulation is the brain’s redundancy in maintain overall homeostasis, with adequate blood flow supply being no exception. 42 Overlapping mechanisms contribute to maintain CBF under challenging circumstances. Therefore, another research challenge when studying CBF control is the overlap and interplay of all mechanisms, and the overall effects on pathway derangements in diseased states. It is very likely that multiple mechanisms act simultaneously and together. It is generally accepted that the myogenic and metabolic mechanisms control CBF and thereby maintain CA homeostasis, which is further modified by the neurogenic mechanism. The molecular processes underlying these mechanisms, and therefore CA, is largely unknown. However, key pathways in endothelial dysfunction and vascular inflammation have been identified, and potential candidates along these pathways will be discussed below.
Endothelial dysfunction and vascular inflammation: vasoactive substances in brain injury
Endothelial dysfunction is characterized by a predominance of a more pro-inflammatory state and reduced vasodilation, and hence reduced CBF. 43 This dysfunction of the endothelial cells in cerebral blood vessels occurs commonly after brain injury, and often leads to decreased CBF to the already ischaemic brain. 44
Endothelial derived vasoactive substances may provide a promising link between neuroinflammation and CA autoregulation disturbances, particularly cerebrovascular pressure reactivity impairment. Figure 2 provides a summary of the various potential pathways involved in CBF control. Importantly, these pathways are complex, and the mediators mentioned below often act in an environmentally dependent fashion, where specific fine-tuning of the cellular environment can alter the function of the mediator. Here, we discuss candidate mediators while acknowledging the complexities and intricacies involved in studying their functions and actions in health and disease.
Figure 2.
Schematic figure (not to scale): under normal healthy conditions, constriction and dilation of the blood vessel are tightly regulated to maintain a constant cerebral blood flow (CBF). Nitric oxide (NO) and prostacyclin induce vasodilation which results in increased cerebral blood volume (CBV). On the other hand, endothelin-1 (ET-1), 20-hydroxyeicosatrienoic-acid (20-HETE), and oxidative stress induce vasoconstriction, which leads to increased CBV. In pathologies such as TBI and SAH, these vasoactive substances that influence vasoconstriction and vasodilation can be up- or down-regulated, and hence CBF may be impacted. The influence of the complement system, nuclear factor kappa B (NF-kB) and the interplay between all the vasoactive substances is largely unknown in brain injury.
Nitric oxide (NO)
Nitric oxide (NO) is the archetypal vasodilator that acts on the smooth muscle cells of cerebral arterial walls. NO is synthesized from the amino acid L-arginine by nitric oxide synthase (NOS), a membrane bound enzyme. Release of NO results in cytosolic calcium release, and causes relaxation of smooth muscle fibres, thereby resulting in vasodilation and increased CBF.45,46 Apart from its vasodilatory function, NO also reduces vascular permeability, platelet aggregation, tissue oxidation and inflammation, cell growth and proliferation and migration. 46 However, there is also literature to suggest NO results in increased blood brain barrier (BBB) permeability. 47 Contradictory findings in the literature48,49 highlight the complexity of NO signalling, and the difficulty in characterising the exact effects of this molecular pathway.
There are three isoforms of NOS: endothelial (eNOS), neuronal (nNOS), and inducible (iNOS, Table 1). nNOS, and eNOS are expressed constitutively in healthy conditions and its production is calcium-dependent, while iNOS is not expressed unless induced by pro-inflammatory cytokines or endotoxins, often in pathology. iNOS production is calcium-independent and is therefore present 100–1000 fold higher than the other NOS-derived isoforms.50,51 In healthy conditions, CBF is likely controlled via the nNOS and eNOS isoform-derived NO pathways. However, in TBI and SAH there is the conceivable added effect of iNOS on CBF control.
Table 1.
A summary of the three different isoforms of NOS, and main functions of the NO derived from this isoform of NOS.
| Isoform of NOS a | Where it is expressed 51 | Main functions of NOS-derived NO |
|---|---|---|
| eNOS | Vascular endotheliumChoroid plexus | Regulation of vascular hemodynamics51,154–156Preserving and maintaining the cerebral microcirculation 156 Inhibiting platelet aggregation, leukocyte adhesion and migration 157 Reducing smooth muscle proliferation 156 |
| nNOS | Neuronal cell bodies | Neurovascular coupling51,156,158,159Neuronal plasticity 160 Memory formation 160 Regulation of CBF 161 Transmission of pain signals 162 Neurotransmitter release 161 |
| iNOS | MacrophagesGlial cellsVascular smooth muscle and endothelial cellsTumour cellsChoroid plexus | Inflammation51,156,163 |
NO has multiple complex roles in normal physiological conditions, and therefore its effects may conceivably be both advantageous and detrimental in pathological states. In traumatic brain injury (TBI), NO accumulates in the brain immediately after injury, seems to be rapidly depleted 30 minutes to 6 hours post-injury, and then gradually accumulates again after 6 hours. 52 Experimental models have demonstrated increased NO concentrations immediately after trauma, measured directly with a NO electrode and indirectly via microdialysate concentrations.53,54 This initial peak is likely the effect of upregulated eNOS and nNOS as iNOS was undetectable. 55 Further exploration using electron paramagnetic resonance spectroscopy in mice brains showed that 90% of the accumulating NO in the brain within the first 10 to 20 minutes after the onset of global cerebral ischaemia was produced by nNOS, while the other 10% was produced via the eNOS. NO produced by nNOS during this time is likely to have adverse effects on outcome, whereas eNOS-produced NO is likely to play an advantageous role by preserving CBF. 52 This dual effect of NO has been seen in ischaemia models,49,56 and studies using NOS inhibitors,52,54 strongly suggesting this action of NO following trauma. As summarised by Cherian et al., the administration of general NOS inhibitors (i.e., not specifically targeting any isoform of NOS) found mixed effects on outcome following trauma in the animal models, however specific inhibition of nNOS is found to be protective, thereby suggesting a detrimental action of nNOS 52 in this initial time-period.
Relative depletion of NO after this initial peak follows for a combination of reasons. Firstly, NOS gene expression can be impaired after trauma, and decreased production of NOS could be because of this oxidative DNA damage. This can recover with time. Additionally, NOS requires 3 co-substrates: L-arginine, oxygen, and nicotinamide adenine dinucleotide phosphate (NADPH). Following trauma, a lack of these necessary substrates is likely to impair NO synthesis. Finally, there could be inhibition of NOS, and inactivation of NOS by free radicals (e.g. superoxide) following trauma. This overall decreased bioavailability of NO (increased metabolism of NO, lack of NOS or likely both) puts the brain at risk of decreased CBF and ischaemia. After 6 hours, NO gradually accumulates and stabilizes. This is likely to be because of iNOS production, which may account for general increase in NO levels during this ‘late’ phase. However, there is some conflicting evidence of the role of iNOS following acute brain injury – whether it is detrimental or neuroprotective following trauma is still unknown, but most models show that the inhibition of iNOS has advantageous effects.52,57
Villalba et al. used rat models to demonstrate that TBI causes increased NO production following injury via an upregulation of iNOS in endothelial cells. 58 This led to enhanced vasodilation and the loss of myogenic tone. The authors postulate that defective myogenic tone may be contributing to the loss of CA following TBI but recognise that it is unclear whether enhanced vasodilation is a protective response, or a deleterious contributor to secondary injury following injury.
NO is a short-lived, highly reactive species but its major products nitrate and nitrite (collectively termed NOx) can readily be measured retrospectively in biological samples as markers of nitric oxide production. A pilot study by Carpenter et al. of 12 patients (11 TBI, one SAH) receiving neurocritical care has shown that as microdialysate NOx increases, glucose increases, lactate decreases, and the L/P ratio decreases. 59 These findings suggest that in human acute brain injury, NO may improve blood supply, delivering more oxygen and glucose to brain tissue, and shifting metabolism towards the TCA cycle. The general temporal pattern was that NOx concentrations within-patient were relatively higher at earlier time-points compared with later time-points, 59 consistent with the findings of Hlatky et al. in TBI, 60 and of Sakowitz et al. in SAH. 61 In the SAH patients studied by Carpenter et al., microdialysis started 2 days post-SAH and continued for 10 days, with NOx concentrations measured in the final 2 days, during which time fluctuations in NOx were followed by a decline to markedly low levels, with unfavourable clinical outcome.
SAH patients experience a decrease in CBF, which may be further exacerbated by NO depletion. NO signalling pathways have been shown to play a significant role in the risk of delayed cerebral ischaemia following SAH. This is of particular importance for the eNOS gene, as some genetic variants influences the patient’s risk of DCI following SAH as lower eNOS activity results in increased risk, 62 showing the protective effect of eNOS in SAH, as in TBI.
Additionally, the functional uncoupling of eNOS from its co-factor tetrahydrobiopterin (BH4) during cerebral ischaemia, and results in the production of superoxide instead of NO. 63 Superoxide then goes on to react with NO to form peroxynitrite – this is detrimental as it not only decreases bioavailability of NO, but peroxynitrite is neurotoxic itself,51,64,65 causing mitochondrial damage, vascular endothelial and smooth muscle cell damage. 66
In TBI and SAH, increased oxidative stress is a significant contributor to the development of secondary injury, and exacerbation of pathology.67,68 Multiple sources of excessive free radical generation include disrupted metabolism and mitochondrial functioning, as well as upregulation of enzymes that produce free radicals, and downregulation of enzymes involved in antioxidant systems. Reactive oxygen species can lead to smooth muscle cell hypertrophy and endothelial apoptosis, which may contribute to dysfunctional CA and CBF maintenance. 69 Oxidative stress has been consistently associated with reduced NO function, and hence reduced endothelium-dependent relaxations. Reactive oxygen species inhibit the vasodilatory functions of NO, 45 causing further decrease in CBF following acute brain injury.
While there is a large body of evidence to implicate the NOS/NO pathway in CA and CBF control after TBI, there is conflicting evidence about the role that each isoform of NOS has to play. Theoretically, it seems that that an upregulation of eNOS, and downregulation of nNOS and iNOS could preserve CBF after injury, and possibly avoid exacerbation of secondary ischaemia.
Prostaglandins
Prostaglandins are a group of lipids that have both homeostatic and pathological functions. Like many inflammatory mediators, prostaglandins can be beneficial and deleterious following injury, but they act primarily as powerful vasodilators (although some animal work suggests prostaglandins may have vasoconstrictory properties as well70–72). These lipids act locally on the smooth muscle cells in the cerebral blood vessels, and also play a role in blood vessel permeability (as pro-inflammatory production of prostaglandins facilitate leukocyte movement from blood vessels to injured tissue). Therefore, it is unsurprising that prostaglandin changes may alter CBF and vascular reactivity. Prostacyclin, an organic compound in the prostaglandin family, has been implicated in mediating autoregulatory responses as it is produced in the walls of cerebral vessels. It not only acts as a potent vasodilator, but also inhibits platelet aggregation and is reduced in the acute period following SAH.73,74 This suggests that its function as a regulator of CBF may be impaired in SAH, implicating this compound’s mechanistic role in CA.
Endothelin-1
Endothelial cells are stimulated to produce endothelin-1 (ET-1) by ischaemic insult and shear stress, 75 causing vasoconstriction. In normal conditions, ET-1 release is tightly controlled and balanced with vasodilatory modulators, and the balance between vasoconstrictors (such as ET-1) and vasodilators (mainly NO) is vital in controlling CBF and endothelial function. 76 However, after brain injury, this balance is disrupted. SAH patients not only have an upregulation of ET-1, 77 but increased expression of the ET-1 receptor on the endothelium of cerebral arteries.78,79 Elevated levels of ET-1 are also seen following TBI 80 as well as after acute ischaemic stroke81,82 where higher ET-1 serum concentrations were associated with more severe brain oedema. 83 The upregulation of ET-1 after brain injury is associated with clinical vasospasm and secondary injury likely because of its efficiency and long-lasting vasoconstriction effects. 84 The source of ET-1 over-production has been investigated, with active astrocytes and endothelial cells being key candidates. 85 Theoretically, treatment aimed at decreasing ET-1 levels would be advantageous in increasing CBF and ameliorating some of ET-1’s harmful effects. A clinical trial in SAH patients looked at the effect of a selective ET-A receptor antagonist, clazosentan. The CONSCIOUS I-trial (Clazosentan to Overcome Neurological Ischaemia and Infarct OccUrring after Subarachnoid haemorrhage) showed a dose-dependent decrease in moderate to severe vasospasm, delayed ischaemic neurological deficits and new infarcts (seen on CT scan).85,86 This implicated ET-1 in the development of vasospasm in SAH. However, the CONSCIOUS II and III trials found no evidence that ET antagonists improved clinically relevant outcomes in SAH patients.87,88 This trial therefore provides evidence against a pure focus on macrovascular vasospasm as the cause of delayed ischaemia and resulting neurological deficit which is as expected: the development of vasospasm, just like the role of ET-1, is complex and involves several pathways, including microvascular thrombosis and brain injury induced at the time of ictus.
20-hydroxyeicosatrienoic-acid (20-HETE)
Vascular smooth muscle cells in cerebral artery walls express cytochrome P enzymes that result in the production of 20-hydroxyeicosatrienoic-acid (20-HETE), and that 20-HETE levels increase in response to elevations in intravascular pressure. 89 20-HETE is a potent vasoconstrictor arachidonic acid that is known to play a role in microvasculature in cerebral circulation, CBF control, inflammatory reactions, and cell proliferation.90–94 Inhibiting 20-HETE has shown advantageous effects in animal models: attenuated 20-HETE levels reduce cerebrovascular damage and improve stoke outcomes after cerebral ischaemia.90,95–97 Thus, it has been suggested that the administration of such an inhibitor could be neuroprotective after TBI and SAH. Lu et al. showed that inhibiting 20-HETE via the administration of a 20-HETE inhibitor, HET0016, protected against brain oedema in a traumatic rat model. Inhibition of 20-HETE also decreased blood brain barrier permeability following trauma. The group suggest that the administration of HET0016 decreased oxidative stress following trauma, and that allowed for the preservation of tight junction proteins and BBB integrity. 98
20-HETE has been implicated in the myogenic response and CBF control89,99 and increased 20-HETE has been associated with increased myogenic response and CA adaptation to hypertension.100–102 Additionally, it has been shown that NO inhibits the cytochrome P enzymes involved in 20-HETE production, and that inhibition of 20-HETE contributes to vasodilation via NO pathways. 103 Hence, this is further evidence that the balance between vascular mediators that promote vasoconstriction and dilation is vital for CA and CBF maintenance. We can also note that vasodilation via NO pathways has both 20-HETE dependent and independent (via guanylate cyclase47,104–107) mechanisms.
Nuclear factor kappa B (NF-kB)
Nuclear factor kappa B (NF-kB) is a transcription factor that regulates the immune response, particularly promotes the production of pro-inflammatory cytokines, chemokines and cell adhesion molecules as part of innate immunity. The function of NF-kB has been explored, with endothelial, glial, and neuronal activation found during cerebral ischaemia.108–110 The exact vascular function of NF-kB in in healthy conditions and following injury, is largely unknown. However, its activation during cerebral ischaemia and the fact that the addition of putative neuroprotective antioxidants inhibits this NF-kB activation (and consequently results in reduced ischaemic neuronal death) suggests that the role of NF-kB following brain injury is deleterious. 111 Interestingly, the fact that antioxidants can be used as NF-kB inhibitors suggests that intermediates of the reactive oxygen pathway are involved in NF-kB activation, possibly via a secondary messenger system.
Endothelial activation of this transcription factor may initiate, and exacerbate the pro-inflammatory cascade involved in the immediate hours following acute brain injury, and hence may play a role in CBF regulation. However, as NF-kB is an inducible transcription factor, it will likely be involved in the upstream CA molecular mechanisms and may have a non-specific inflammatory role in CBF regulation.
Complement system
The complement system is part of the innate immune system and is a key pathway contributing to neuroinflammation and secondary injury following TBI.112–114 The complement cascade is controlled by multiple mediators, and the lack of this control leads to exacerbated neuropathology after trauma. 115
There is evidence that complement plays a crucial role in the activation of microglia in the early stages of the immune response, as microglia express receptors for various complement proteins, including C1q and C3 cleavage products.116,117 During pathology, the C1q protein promotes the shift of microglia phenotype to M1, the pro-inflammatory phenotype involved with clearing cellular debris and foreign antigens but can also be harmful to healthy cells and cause further damage. 118 Complement proteins C3a and C5a bind to their receptors and induce neuronal and glial apoptosis, as well as increase BBB leakage. 119 With BBB disruption, a further influx of complement proteins exacerbates the inflammatory response. 120 The terminal complement complex (TCC) is a downstream protein indicative of persistent and irreversible complement activation, and this protein further promotes neuronal lysis. Uncontrollable activation of the complement system therefore results in oxidative stress and free radical production, cell signalling disruption and further cell damage and apoptosis, thereby contributing significantly to secondary insult following brain injury.
The complement system has three different activation pathways: the classical, the alternative, and the lectin pathway. 121 All pathways are triggered by complement initiator proteins binding to antibodies on antigens, or damaged cells. The activation of all pathways can be triggered by foreign antigen cells, as well as damaged endogenous cells– pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) respectively. The pathways are activated by different initiator molecules, and have different effector molecules, but these pathways all converge in that they all form C3 and C5 convertase, and subsequently activate the terminal complement pathway. 119
There is evidence to suggest that all three pathways are active after brain injury. The key role of specific pathways is somewhat unknown and can be difficult to study considering that the pathways are intertwined and converge at common proteins. Animal studies have attempted to investigate the role of the three activation pathways. Mice lacking fB, a fundamental mediator in the alternative pathway (and hence whose alternative pathways are non-functional) has reduced neuronal loss, upregulation of anti-apoptotic proteins, and down regulation of anti-apoptotic receptors. 122 In a similar study, alternative pathway inhibition via fB also showed reduced neuronal loss, decreased inflammatory response, and upregulated neuroprotection pathways, however there was no improvement in neurological functioning in these mice models.123,124
Animal model studies of TBI have investigated mice deficient for C4 (a protein involved in both the classical and lectin pathways) and found reduced tissue damage and motor deficits compared to wild type mice after controlled cortical injury. A study found that early administration of C1 inhibitor (functionally inhibiting both the classical and lectin pathways) allowed for preserved cognitive functioning and smaller contusion size, whereas delayed administration led to a reduction in motor dysfunction but had no effect on cognition or contusion size. 125 Attempts at isolating the effects of these pathways further have been made. Substantial involvement of the classical pathway in TBI pathology is questioned, as mice deficient for C1q (a classical initiator) showed similar neurological deficits and lesion sizes to controls. 126
There is a large body of evidence that implicates the lectin pathway in acquired brain injury, and hence potential analytes of this pathway could act as key biomarkers of CBF disturbance (Figure 3). Animal models have shown that activation of the lectin pathway early after TBI may have neuroprotective effects as mice without mannose binding lectin (MBL, an initiator protein) had greater neurological deficits and increased neurodegeneration in the early phase after cortical injury compared to controls. 127 Notably, MBL has been found to be selectively deposited on the ischaemic endothelium as early as 30 minutes after focal cerebral ischaemia in mice. 128 Five weeks post injury, mice models deficient in MBL had reduced neuronal loss, 129 suggesting lectin pathway activity at the later stages after TBI are detrimental. The detrimental effects of lectin pathway activation are largely supported with evidence in cerebral ischaemia, 128 in particular TBI, 130 stroke, 131 and SAH. 132
Figure 3.
Evidence suggesting full and persistent activation of the lectin complement pathway after brain injury nominates potential candidate markers that could be associated with cerebral autoregulatory dysfunction.
A study by De Blasio et al. investigated lectin complement pathway activation in early (within 12 hours from injury) and late (12 hours to 5 days after injury) TBI patients. Immunofluorescence and confocal imaging analysis showed that lectin initiator molecules (MBL and ficolins), key enzymes (MBL-associated serine proteases, MASPs) and downstream components (C3 fragments and TCC) were elevated in TBI contusions compared to non-TBI contusions. These lectin complement proteins were found in the blood vessels and brain parenchyma of the TBI patients, and higher levels of MASP-2 were associated with altered pupil reactivity after traumatic SAH, and hence more severe clinical conditions. 133 This finding is supported,113,130,134 with Burk et al. showing that serum levels of C3a and C5a of polytrauma patients correlate with injury severity and survival. This group further suggested that complement is rapidly activated and dysfunctional, thereby resulting in “complementopathy” after trauma. 113
Lectin complement is not unique to TBI, however, De Blasio et al.’s evidence suggests that TBI favours full and persistent activation of the complement system, and its localisation to blood vessels as well as brain tissue of contusions suggests its contribution to secondary brain injury in acute brain damage pathophysiology. This study concluded that this complement activation depends on the lectin activation pathway with possible amplification by the alternative pathway; this activation is persistent up until 5 days post injury and is associated with injury severity. 133
Measuring these candidate markers
An ideal biomarker of CA ‘health’ would be a specific inflammatory marker that would be associated with the status of CA (i.e., reflecting whether CA is globally intact or impaired) of the patient, thus informing if CPP management efforts are at all likely to achieve improvement in CA. This candidate biomarker would need to be easily measured, and the levels rapidly quantified.
Inherent properties of the candidate marker would need to be considered. The molecular weight, half-life of the candidate marker, its stability and degradation at room temperature, and how the sample processing would affect the marker would affect the method of analysis and measurement. This is of particular concern for the NO pathway. NO production from the individual isoforms of NO synthase are difficult to study and there is currently no method to measure individual isoform-derived NO in human samples. Studies to date have looked at the overall NO levels via an invasive NO sensor (such as NO-selective electrodes used in-vivo in animal studies) or have studied NO metabolism products – nitrite and nitrate (measured ex-vivo in biofluids from patients or animals). Both methods do not give an indication of the source of the NO. Therefore, the study of NOS isoforms has come from animal models, via the use of specific NOS isoform inhibitors. This gives an indication of the effect of the specific NO in a working system, however the translational ability of this research to humans must be considered.
Additionally, the human sample that will be analysed for the candidate marker must be considered. Each sample: blood, cerebrospinal fluid (CSF) and microdialysis fluid pose challenges for methods of analysis. Broadly, a plasma or serum sample would have the added influence of systemic function, and its analytes would not be directly cerebral. Arterio-jugular venous differences have been suggested as a means of determining the net production of mediators across the cerebrovascular bed, however, the small absolute difference between the samples makes identification of a consistent difference difficult. 135 Ventricular CSF is valuable, as it is central nervous system-specific, but in general is not readily accessible in TBI patients as the use of extra-ventricular drains (EVDs) as part of clinical management depends on the local practice and varies between countries and trauma centres. Microdialysis monitoring has been shown to be useful in clinical practice because it offers insight into brain metabolism. In addition, it samples analytes directly from the extracellular fluid and hence may offer insight into processes occurring directly in the brain. However, the analysis of microdialysate is challenging as the catheter membrane filter only allows retrieval of substances with a molecular weight less than 100 kDa. Furthermore, the sample volume yield is low which often requires pooling of samples for meaningful analysis, depending on the analyte(s). This then decreases the temporal information we can investigate, regarding the patient’s inflammatory profile.
An enzyme-linked immunosorbent assay (ELISA) would be ideal for the quantification of candidate markers in the blood, CSF or microdialysate of human patients as it is relatively quick, affordable, and easy to use. The detection range, and degree of specificity varies from plate to plate, however a high sensitivity and specificity is generally considered an advantage of this method. Furthermore, a multiplexing method (such as Luminex®), would allow for the simultaneous investigation of multiple potential biomarkers along a candidate pathway – where the analysis of upstream activation markers, intermediate effector markers, as well as terminal or downstream candidates can be explored in one sample run. This would provide additional information about the pathway, such as extent of activation in pathology. However, ELISA and Luminex assays are offline techniques that require several hours’ analysis time in a laboratory with specialised equipment, so cannot be used for assessing patients in near-real-time that would be necessary for implementing individualised therapy. Thus, point-of-care rapid assays would be highly desirable, with potential for patient benefit.
Assessing cerebral autoregulation in humans
Human CA assessment is difficult, partly because it is a concept and not a directly quantifiable phenomenon. 136 Assessment of autoregulation is traditionally classified into two methods - static and dynamic testing. Loosely speaking, this division is dependent on whether assessment is based on the degree of suppression of the blood pressure driven changes in cerebral blood flow, or the speed of engagement of that mechanism. Static readings can be achieved by measuring changes in CBF (or CBFV, cerebral blood flow velocity) in response to induced step changes in MAP (measurements taken before and after the change in MAP has stabilised). These can be obtained via the administration of medication or a shift in blood volume. 136 Static testing provides direct information about the range of CPP in which CA is active and can be used to recreate the classic autoregulation curve. 137 Dynamic CA testing, on the other hand, refers to the rapid phase of changes in MAP, where the CBF (or surrogate thereof) response time gives us an indication of CA.138,139 As this method of CA assessment does not require a new steady state to be achieved (thus allowing rapid and readily repeated assessment), it may be more clinically useful. MAP pressure changes can be induced or spontaneous, with induced changes including the thigh cuff release, 140 Valsalva manoeuvre, 141 tilt test 142 or transient hyperaemic response test. 143 Assessing cerebral autoregulation with spontaneous fluctuations in MAP can be done in the time and frequency domain. Spontaneous MAP fluctuations are inherently non-stimulatory and therefore provide autoregulatory information without the additional risks of static testing, which makes it applicable to a wider patient population. 144
Most accessible CA testing involves a surrogate measure of CBF or cerebral blood volume (CBV) changes, which can be invasive or non-invasive. Invasive surrogates include brain tissue oxygenation monitoring (CBF surrogate), and ICP monitoring (CBV surrogate), where signals from these monitoring methods can be correlated with MAP, and the resulting correlation coefficient provides autoregulatory information. 20 PRx, the correlation index between MAP and ICP, is the most widely used index of pressure reactivity and can be calculated continuously and in real time at the patient bedside. 20 The current gold-standard of non-invasive CBF measurement is the transcranial Doppler (TCD)-measured CBFV, with Mx being the correlation coefficient between MAP and CBFV.140,145 The use of Near Infrared Spectroscopy (NIRS) in non-invasive CA assessment is gaining popularity as it may provide an opportunity for easy, long-term continuous monitoring. 146
It is of great importance to consider the method of CA assessment when attempting to relate CA status and the inflammatory cascade. Biomarker levels may have better associations with certain CA indices and not others, and in order to identify key molecular pathways involved in CA disturbance, methods of assessment need to be considered. Furthermore, the clinical impact of individualised management of blood pressure, or cerebral perfusion pressure based on the continuous monitoring of CA, 147 is likely to be much improved if augmented by better understanding of those pathways.
Applicability and transferability to other pathological states in humans
CBF disturbance and the loss of CA is a common cascade following several neurological pathologies such as stoke,148,149 meningitis,150,151 and intracerebral haemorrhage,152,153 as well as non-neurological originating pathologies like metabolic disease. 27 Hence, work on identifying the molecular pathways underpinning CA and CBF control, and how derangements in these pathways manifest clinically, has wider applicability and treatment potential.
While each disease manifests in a unique way in every patient, the molecular underpinnings of this common pathway and CBF control may be maintained across diseases. This primary research into vascular inflammation and mediators of CBF control could identify the underlying molecular pathways, and targets to control these pathways will work towards avoiding and/or ameliorating CA dysfunction in the clinical setting. This may have a positive effect on mortality and outcome for many pathological states and neurological diseases. While the primary pathways may be common for these pathologies where CA is disturbed, it is important to note that the pathological mechanisms (especially of primary and secondary brain injury) and consequences of proinflammatory states differ. Hence, it is likely that specific biomarkers will be elevated in some states, and not others. Therefore, fundamental explorations of CBF control may be applicable and transferable, but development of specific biomarkers indicative of CA status of the patient, may need more fine-tuning to the disease, mechanism of pathology, and the individual.
Conclusion: comments on future work and clinical utility
Endothelial dysfunction and vascular inflammation offer a plausible link to CA disturbances following both SAH and TBI, and, in principle, a promising avenue for therapy. This review has discussed candidate biomarkers that provide the highest yield inflammatory cascades involved in CBF maintenance. While many of these pathways overlap and influence each other, candidate markers of these pathways may offer insight into the molecular underpinnings of CBF disturbances in brain injury. Clinical trials, such as the ongoing investigations into optimal CPP targets, could incorporate the measurement of suggested candidate markers. In doing so, such studies could connect candidate inflammatory pathways with CBF disturbances, and addressing these disturbances may improve treatment of acute brain injury patients.
The easy and robust measurement of these candidate markers will depend on many factors, and future work could aim at exploring these pathways to identify the most influential ones in CA. Considering the difficulties of measuring NO derived from the individual isoforms of NOS, the complement pathway (particularly lectin activation) may offer the most promise.
In conclusion, this review describes candidate biomarkers of CA and mechanisms at which future work can be directed. Once the molecular underpinnings of CBF maintenance have been identified, further studies into targeting these pathways, and hence attenuating or possibly preventing CA impairment after acute injury can be conducted, with the potential to improve patient management and outcome.
Footnotes
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: Medical Research Council (Grant no. G1002277 ID98489) and National Institute for Health and Care Research Biomedical Research Centre, Cambridge (Neuroscience Theme; Brain Injury and Repair Theme). Authors’ support: CAS– Patrick & Margaret Flanagan Skye Cambridge Trust Scholarship; KLHC–National Institute for Health and Care Research Biomedical Research Centre, Cambridge (Neuroscience Theme; Brain Injury and Repair Theme), NIHR Invention for Innovation (i4i), and NIHR Brain Injury MedTech Co-operative; PJH–National Institute for Health and Care Research (Professorship, Biomedical Research Centre, Brain Injury MedTech Co-operative, Senior Investigator Award and the Royal College of Surgeons of England, and NIHR Invention for Innovation (i4i). PS–nothing to disclose; AH–Medical Research Council/Royal College of Surgeons of England Clinical Research Training Fellowship (Grant no. G0802251), the NIHR Biomedical Research Centre and the NIHR Brain Injury MedTech Co-operative; The views expressed are those of the authors and are not necessarily those of the NIHR, the Department of Health and Social Care or of any of the other funding bodies.
The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: P.J.H. is a director of Technicam (Newton Abbot, UK), the manufacturer of the cranial access device used in several of the microdialysis studies cited in this article. P.S. has a financial interest in part of the licensing fee for ICM+ software for brain monitoring (www.neurosurg.cam.ac.uk/icmplus) which is licensed by the University of Cambridge (Cambridge Enterprise Ltd). The other authors have no conflict of interests to declare.
ORCID iDs: Claudia A Smith https://orcid.org/0000-0003-3643-6191
Adel Helmy https://orcid.org/0000-0002-0531-0556
References
- 1.Rangel-Castilla L, Gasco J, Nauta HJW, et al. Cerebral pressure autoregulation in traumatic brain injury. Neurosurg Focus 2008; 25: E7. [DOI] [PubMed] [Google Scholar]
- 2.Miller JD, Stanek A, Langfitt TW. Concepts of cerebral perfusion pressure and vascular compression during intracranial hypertension. 1972; pp : 411–432. [DOI] [PubMed]
- 3.Bouma GJ, Muizelaar JP, Choi SC, et al. Cerebral circulation and metabolism after severe traumatic brain injury: the elusive role of ischemia. J Neurosurg 1991; 75: 685–693. [DOI] [PubMed] [Google Scholar]
- 4.Budohoski KP, Czosnyka M, de Riva N, et al. The relationship between cerebral blood flow autoregulation and cerebrovascular pressure reactivity after traumatic brain injury. Neurosurgery 2012; 71: 652–661. [DOI] [PubMed] [Google Scholar]
- 5.Czosnyka M, Smielewski P, Piechnik S, et al. Cerebral autoregulation following head injury. J Neurosurg 2001; 95: 756–763. [DOI] [PubMed] [Google Scholar]
- 6.Panerai RB, Kerins V, Fan L, et al. Association between dynamic cerebral autoregulation and mortality in severe head injury. Br J Neurosurg 2004; 18: 471–479. [DOI] [PubMed] [Google Scholar]
- 7.Donnelly J, Czosnyka M, Adams H, et al. Twenty-five years of intracranial pressure monitoring after severe traumatic brain injury: a retrospective, single-center analysis. Neurosurg 2019; 85: E75–E82. [DOI] [PubMed] [Google Scholar]
- 8.Morganti-Kossmann MC, Rancan M, Stahel PF, et al. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care 2002; 8: 101–105. [DOI] [PubMed] [Google Scholar]
- 9.Simon DW, McGeachy MJ, Bayır H, et al. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol 2017; 13: 572–572. [DOI] [PubMed] [Google Scholar]
- 10.Finnie JW. Neuroinflammation: beneficial and detrimental effects after traumatic brain injury. Inflammopharmacol 2013; 21: 309–320. [DOI] [PubMed] [Google Scholar]
- 11.Myer DJ, Gurkoff GG, Lee SM, et al. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain 2006; 129: 2761–2772. [DOI] [PubMed] [Google Scholar]
- 12.Schmidt OI, Infanger M, Heyde CE, et al. The role of neuroinflammation in traumatic brain injury. Eur J Trauma 2004; 30: 135–149. [Google Scholar]
- 13.Overgaard J, Tweed WA. Cerebral circulation after head injury. Part 1: cerebral blood flow and its regulation after closed head injury with emphasis on clinical correlations. J Neurosurg 1974; 41: 531–541. [DOI] [PubMed] [Google Scholar]
- 14.Rodríguez-Baeza A, Reina-De La Torre F, Poca A, et al. Morphological features in human cortical brain microvessels after head injury: a three-dimensional and immunocytochemical study. Anat Rec 2003; 273A: 583–593. [DOI] [PubMed] [Google Scholar]
- 15.Zeiler FA, Thelin EP, Donnelly J, et al. Genetic drivers of cerebral blood flow dysfunction in TBI: a speculative synthesis. Nat Rev Neurol 2019; 15: 25–39. [DOI] [PubMed] [Google Scholar]
- 16.Griessenauer CJ, Starke RM, Foreman PM, et al. Associations between endothelin polymorphisms and aneurysmal subarachnoid hemorrhage, clinical vasospasm, delayed cerebral ischemia, and functional outcome. J Neurosurg 2018; 128: 1311–1317. [DOI] [PubMed] [Google Scholar]
- 17.Griessenauer CJ, Tubbs RS, Foreman PM, et al. Associations of renin-angiotensin system genetic polymorphisms and clinical course after aneurysmal subarachnoid hemorrhage. J Neurosurg 2017; 126: 1585–1597. [DOI] [PubMed] [Google Scholar]
- 18.Dardiotis E, Paterakis K, Siokas V, et al. Effect of angiotensin-converting enzyme tag single nucleotide polymorphisms on the outcome of patients with traumatic brain injury. Pharmacogenet Genomics 2015; 25: 485–490. [DOI] [PubMed] [Google Scholar]
- 19.Bayliss WM. On the local reactions of the arterial wall to changes of internal pressure. J Physiol 1902; 28: 220–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous assessment of the cerebral vasomotor reactivity in head injury. Neurosurgery 1997; 41: 11–19. [DOI] [PubMed] [Google Scholar]
- 21.Sorrentino E, Diedler J, Kasprowicz M, et al. Critical thresholds for cerebrovascular reactivity after traumatic brain injury. Neurocrit Care 2012; 16: 258–266. [DOI] [PubMed] [Google Scholar]
- 22.Fog M. The relationship between the blood pressure and the tonic regulation of the pial arteries. J Neurol Neurosurg Psychiatry 1938; 1: 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kontos HA, Wei EP, Navari RM, et al. Responses of cerebral arteries and arterioles to acute hypotension and hypertension. Am J Physiol Heart Circ Physiol 1978; 234: H371–H383. [DOI] [PubMed] [Google Scholar]
- 24.Halpern W, Osol G. Influence of transmural pressure on myogenic responses of isolated cerebral arteries of the rat. Ann Biomed Eng 1985; 13: 287–293. [DOI] [PubMed] [Google Scholar]
- 25.Osol G, Halpern W. Myogenic properties of cerebral blood vessels from normotensive and hypertensive rats. Am J Physiol Heart Circ Physiol 1985; 249: H914–H921. [DOI] [PubMed] [Google Scholar]
- 26.Knot HJ, Nelson MT. Regulation of arterial diameter and wall [Ca2+] in cerebral arteries of rat by membrane potential and intravascular pressure. J Physiol 1998; 508: 199–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Coucha M, Abdelsaid M, Ward R, et al. Impact of metabolic diseases on cerebral circulation: structural and functional consequences. Compr Physiol 2018; 8: 773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Webb RC. Smooth muscle contraction and relaxation. Am J Physiol – Adv Physiol Educ 2003. ; 27: 201–206. [DOI] [PubMed] [Google Scholar]
- 29.Phillips AA, Chan FH, Zheng MMZ, et al. Neurovascular coupling in humans: physiology, methodological advances and clinical implications. J Cereb Blood Flow Metab 2016; 36: 647–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Murkin JM. Cerebral autoregulation: the role of CO2 in metabolic homeostasis. Semin Cardiothorac Vasc Anesth 2007. ; 11: 269–273. [DOI] [PubMed] [Google Scholar]
- 31.Faraci FM, Taugher RJ, Lynch C, et al. Acid-Sensing ion channels: novel mediators of cerebral vascular responses. Circ Res 2019; 125: 907–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Okamoto H, Hudetz AG, Roman RJ, et al. Neuronal NOS-derived NO plays permissive role in cerebral blood flow response to hypercapnia. Am J Physiol Heart Circ Physiol 1997; 272: H559–H566. [DOI] [PubMed] [Google Scholar]
- 33.Wang Q, Pelligrino DA, Baughman VL, et al. The role of neuronal nitric oxide synthase in regulation of cerebral blood flow in normocapnia and hypercapnia in rats. J Cereb Blood Flow Metab 1995; 15: 774–778. [DOI] [PubMed] [Google Scholar]
- 34.Wang Q, Pelligrino DA, Koenig HM, et al. The role of endothelium and nitric oxide in rat pial arteriolar dilatory responses to CO2 in vivo. J Cereb Blood Flow Metab 1994; 14: 944–951. [DOI] [PubMed] [Google Scholar]
- 35.Iadecola C, Yang G, Xu S. 7-Nitroindazole attenuates vasodilation from cerebellar parallel fiber stimulation but not acetylcholine. Am J Physiol Regul Integr Comp Physiol 1996; 270 [DOI] [PubMed] [Google Scholar]
- 36.Silverman A, Petersen NH. Physiology, cerebral autoregulation. Treasure Island, FL: StatPearls. 2020. [PubMed]
- 37.Hosford PS, Gourine AV. What is the key mediator of the neurovascular coupling response? Neurosci Biobehav Rev 2019; 96: 174–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Attwell D, Buchan AM, Charpak S, et al. Glial and neuronal control of brain blood flow. Nature 2010; 468: 232–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wei EP, Raper AJ, Kontos HA, et al. Determinants of response of pial arteries to norepinephrine and sympathetic nerve stimulation. Stroke 1975; 6: 654–658. [DOI] [PubMed] [Google Scholar]
- 40.Lee TJ, Hume WR, Su C, et al. Neurogenic vasodilation of cat cerebral arteries. Circ Res 1978; 42: 535–542. [DOI] [PubMed] [Google Scholar]
- 41.Mishra A, Reynolds JP, Chen Y, et al. Mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat Neurosci 2016; 19: 1619–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Claassen JAHR, Thijssen DHJ, Panerai RB, et al. Regulation of cerebral blood flow in humans: physiology and clinical implications of autoregulation. Physiol Rev 2021; 101: 1487–1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Widlansky ME, Gokce N, Keaney JF, et al. The clinical implications of endothelial dysfunction. J Am Coll Cardiol 2003; 42: 1149–1160. [DOI] [PubMed] [Google Scholar]
- 44.Ierssel S, Conraads VM, Craenenbroeck EM, et al. Endothelial dysfunction in acute brain injury and the development of cerebral ischemia. Crit Care 2015; 19: P443. [DOI] [PubMed] [Google Scholar]
- 45.Vanhoutte PM, Shimokawa H, Tang EHC, et al. Endothelial dysfunction and vascular disease. Acta Physiologica 2009; 196: 193–222. [DOI] [PubMed] [Google Scholar]
- 46.Esper RJ, Nordaby RA, Vilariño JO, et al. Endothelial dysfunction: a comprehensive appraisal. Cardiovasc Diabetol 2006; 5: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mayhan WG. VEGF increases permeability of the blood-brain barrier via a nitric oxide synthase/cGMP-dependent pathway. Am J Physiol Cell Physiol 1999; 276: C1148–C1153. [DOI] [PubMed] [Google Scholar]
- 48.Keynes R, Garthwaite J. Nitric oxide and its role in ischaemic brain injury. CMM 2004; 4: 179–191. [DOI] [PubMed] [Google Scholar]
- 49.Iadecola C. Bright and dark sides of nitric oxide in ischemic brain injury. Trends Neurosci 1997; 20: 132–139. [DOI] [PubMed] [Google Scholar]
- 50.Pautz A, Art J, Hahn S, et al. Regulation of the expression of inducible nitric oxide synthase. Nitric Oxide 2010; 23: 75–93. [DOI] [PubMed] [Google Scholar]
- 51.Garry PS, Ezra M, Rowland MJ, et al. The role of the nitric oxide pathway in brain injury and its treatment – from bench to bedside. Exp Neurol 2015; 263: 235–243. [DOI] [PubMed] [Google Scholar]
- 52.Cherian L, Hlatky R, Robertson CS. Nitric oxide in traumatic brain injury. Brain Pathology 2004; 14: 195–201. In: [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sakamoto K-I, Fujisawa H, Koizumi H, et al. Effects of mild hypothermia on nitric oxide synthesis following contusion trauma in the rat. J Neurotrauma 1997; 14: 349–353. [DOI] [PubMed] [Google Scholar]
- 54.Cherian L, Goodman JC, Robertson CS. Brain nitric oxide changes after controlled cortical impact injury in rats. Crit Care Med 1999; 27: 52A. [DOI] [PubMed] [Google Scholar]
- 55.Wada K, Chatzipanteli K, Kraydieh S, et al. Inducible nitric oxide synthase expression after traumatic brain injury and neuroprotection with aminoguanidine treatment in rats. Neurosurgery 1998; 43: 1427–1436. [DOI] [PubMed] [Google Scholar]
- 56.Dalkara T, Yoshida T, Irikura K, et al. Dual role of nitric oxide in focal cerebral ischemia. Neuropharmacology 1994; 33: 1447–1452. [DOI] [PubMed] [Google Scholar]
- 57.Sinz EH, Kochanek PM, Dixon CE, et al. Inducible nitric oxide synthase is an endogenous neuroprotectant after traumatic brain injury in rats and mice. J Clin Invest 1999; 104: 647–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Villalba N, Sonkusare SK, Longden TA, et al. Traumatic brain injury disrupts cerebrovascular tone through endothelial inducible nitric oxide synthase expression and nitric oxide gain of function. J Am Heart Assoc 2014; 3: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Carpenter KLH, Timofeev I, Al-Rawi PG, et al. Nitric oxide in acute brain injury: a pilot study of NOx concentrations in human brain microdialysates and their relationship with energy metabolism. Acta Neurochir Suppl (Wien) 2008; 102: 207–213. [DOI] [PubMed] [Google Scholar]
- 60.Hlatky R, Goodman JC, Valadka AB, et al. Role of nitric oxide in cerebral blood flow abnormalities after traumatic brain injury. J Cereb Blood Flow Metab 2003; 23: 582–588. [DOI] [PubMed] [Google Scholar]
- 61.Sakowitz OW, Wolfrum S, Sarrafzadeh AS, et al. Temporal profiles of extracellular nitric oxide metabolites following aneurysmal subarachnoid hemorrhage. Acta Neurochir Suppl 2002; 81: 351–354. [DOI] [PubMed] [Google Scholar]
- 62.Vellimana AK, Milner E, Azad TD, et al. Endothelial nitric oxide synthase mediates endogenous protection against subarachnoid hemorrhage-induced cerebral vasospasm. Stroke 2011; 42: 776–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Stuehr D, Pou S, Rosen GM. Oxygen reduction by nitric-oxide synthases. J Biol Chem 2001; 276: 14533–14536. [DOI] [PubMed] [Google Scholar]
- 64.Drexler H, Hornig B. Endothelial dysfunction in human disease. J Mol Cell Cardiol 1999; 31: 51–60. [DOI] [PubMed] [Google Scholar]
- 65.Wink DA, Hanbauer I, Krishna MC, et al. Nitric oxide protects against cellular damage and cytotoxicity from reactive oxygen species. Proc Natl Acad Sci USA 1993; 90: 9813–9817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Moro MA, Almeida A, Bolaños JP, et al. Mitochondrial respiratory chain and free radical generation in stroke. Free Radic Biol Med 2005; 39: 1291–1304. [DOI] [PubMed] [Google Scholar]
- 67.Rodriguez-Rodriguez A, Egea-Guerrero J, Murillo-Cabezas F, et al. Oxidative stress in traumatic brain injury. CMC 2014; 21: 1201–1211. [DOI] [PubMed] [Google Scholar]
- 68.Jelinek M, Jurajda M, Duris K. The role of oxidative stress in early brain injury after subarachnoid hemorrhage. Oxid Med Cell Longev 2020; 2020: 1–9. [Google Scholar]
- 69.Ayer RE, Zhang JH. Oxidative stress in subarachnoid haemorrhage: significance in acute brain injury and vasospasm . Vienna: Springer, 2008, pp. 33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yamamoto YL, Feindel W, Wolfe LS, et al. Experimental vasoconstriction of cerebral arteries by prostaglandins. J Neurosurg 1972; 37: 385–397. [DOI] [PubMed] [Google Scholar]
- 71.Lang SA, Maron MB. Role of prostaglandins in blood-induced vasoconstriction of canine cerebral arteries. J Cereb Blood Flow Metab 1988; 8: 109–115. [DOI] [PubMed] [Google Scholar]
- 72.Dabertrand F, Hannah RM, Pearson JM, et al. Prostaglandin E2, a postulated astrocyte-derived neurovascular coupling agent, constricts rather than dilates parenchymal arterioles. J Cereb Blood Flow Metab 2013; 33: 479–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gaetani P, Baena RR, Silvani V, et al. Prostacyclin and vasospasm in subarachnoid hemorrhage from ruptured intracranial aneurysm: a preliminary clinical study. Acta Neurol Scand 2009; 73: 33–38. [DOI] [PubMed] [Google Scholar]
- 74.Budohoski KP, Czosnyka M, Kirkpatrick PJ, et al. Clinical relevance of cerebral autoregulation following subarachnoid haemorrhage. Nat Rev Neurol 2013; 9: 152–163. [DOI] [PubMed] [Google Scholar]
- 75.Suhardja A. Mechanisms of disease: Roles of nitric oxide and endothelin-1 in delayed cerebral vasospasm produced by aneurysmal subarachnoid hemorrhage. Nat Rev Cardiol 2004; 1: 110–116. [DOI] [PubMed] [Google Scholar]
- 76.Bourque SL, Davidge ST, Adams MA. The interaction between endothelin-1 and nitric oxide in the vasculature: new perspectives. Am J Physiol Regul Integr Comp Physiol 2011; 300: R1288–R1295. [DOI] [PubMed] [Google Scholar]
- 77.Suzuki K, Meguro K, Sakurai T, et al. Endothelin-1 concentration increases in the cerebrospinal fluid in cerebral vasospasm caused by subarachnoid hemorrhage. Surg Neurol 2000; 53: 131–135. [DOI] [PubMed] [Google Scholar]
- 78.Masaoka H, Suzuki R, Hirata Y, et al. Raised plasma endothelin in aneurysmal subarachnoid haemorrhage. The Lancet 1989; 334: 1402. [DOI] [PubMed] [Google Scholar]
- 79.Hino A, Tokuyama Y, Kobayashi M, et al. Increased expression of endothelin B receptor mRNA following subarachnoid hemorrhage in monkeys. J Cereb Blood Flow Metab 1996; 16: 688–697. [DOI] [PubMed] [Google Scholar]
- 80.Armstead WM, Kreipke CW. Endothelin-1 is upregulated after traumatic brain injury: a cross-species, cross-model analysis. Neurol Res 2011; 33: 133–136. [DOI] [PubMed] [Google Scholar]
- 81.Sapira V, Cojocaru IM, Lilios G, et al. Study of endothelin-1 in acute ischemic stroke. Rom J Intern Med 2010; 48 [PubMed] [Google Scholar]
- 82.Alioglu Z, Örem A, Bülbül I, et al. Evaluation of plasma endothelin-1 levels in patients with cerebral infarction. Angiology 2002; 53: 77–82. [DOI] [PubMed] [Google Scholar]
- 83.Moldes O, Sobrino T, Millán M, et al. High serum levels of endothelin-1 predict severe cerebral edema in patients with acute ischemic stroke treated with t-PA. Stroke 2008; 39: 2006–2010. [DOI] [PubMed] [Google Scholar]
- 84.Görlach C, Hortobágyi T, Hortobágyi S, et al. Inhibition of endothelin-1 by the competitive ETA receptor antagonist Ro 61-1790 reduces lesion volume after cold injury in the rat. Pflugers Arch 2001; 441: 844–849. [DOI] [PubMed] [Google Scholar]
- 85.Hostenbach S, D’haeseleer M, Kooijman R, et al. J. The pathophysiological role of astrocytic endothelin-1. Prog Neurobiol 2016; 144: 88–102. [DOI] [PubMed] [Google Scholar]
- 86.Macdonald RL, Kassell NF, Mayer S, et al. 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. [DOI] [PubMed] [Google Scholar]
- 87.MacDonald RL, Higashida RT, Keller E, et al. Randomized trial of clazosentan in patients with aneurysmal subarachnoid hemorrhage undergoing endovascular coiling. Stroke 2012; 43: 1463–1469. [DOI] [PubMed] [Google Scholar]
- 88.Macdonald RL, Higashida RT, Keller E, 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. [DOI] [PubMed] [Google Scholar]
- 89.Gebremedhin D, Lange AR, Lowry TF, et al. Production of 20-HETE and its role in autoregulation of cerebral blood flow. Circ Res 2000; 87: 60–65. [DOI] [PubMed] [Google Scholar]
- 90.Zhu J, Wang B, LJ, Armstrong JS, et al. Additive neuroprotection of a 20-HETE inhibitor with delayed therapeutic hypothermia after Hypoxia-Ischemia in neonatal piglets. Dev Neurosci 2015; 37: 376–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Garcia V, Cheng J, Weidenhammer A, et al. Androgen-induced hypertension in angiotensinogen deficient mice: Role of 20-HETE and EETS. Prostaglandins Other Lipid Mediat 2015; 116-117: 124–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Fordsmann JC, Ko RWY, Choi HB, et al. Increased 20-HETE synthesis explains reduced cerebral blood flow but not impaired neurovascular coupling after cortical spreading depression in rat cerebral cortex. J Neurosci 2013; 33: 2562–2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yang ZJ, Carter EL, Kibler KK, et al. Attenuation of neonatal ischemic brain damage using a 20-HETE synthesis inhibitor. J Neurochem 2012; 121: 168–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Harder DR, Gebremedhin D, Narayanan J, et al. Formation and action of a P-450 4A metabolite of arachidonic acid in cat cerebral microvessels. Am J Physiol Heart Circ Physiol 1994; 266: H2098–H2107. [DOI] [PubMed] [Google Scholar]
- 95.Poloyac SM, Zhang Y, Bies RR, et al. Protective effect of the 20-HETE inhibitor HET0016 on brain damage after temporary focal ischemia. J Cereb Blood Flow Metab 2006; 26: 1551–1561. [DOI] [PubMed] [Google Scholar]
- 96.Renic M, Kumar SN, Gebremedhin D, et al. Protective effect of 20-HETE inhibition in a model of oxygen-glucose deprivation in hippocampal slice cultures. Am J Physiol Heart Circ Physiol 2012; 302: H1285–H1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Renic M, Klaus JA, Omura T, et al. Effect of 20-HETE inhibition on infarct volume and cerebral blood flow after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab 2009; 29: 629–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lu L, Wang M, Wei X, et al. 20-HETE inhibition by HET0016 decreases the blood-brain barrier permeability and brain edema after traumatic brain injury. Front Aging Neurosci 2018; 10: 207. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 99.Harder DR, Narayanan J, Gebremedhin D. Pressure-induced myogenic tone and role of 20-HETE in mediating autoregulation of cerebral blood flow. Am J Physiol Heart Circ Physiol 2011; 300: H1557–H1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Toth P, Tucsek Z, Tarantini S, et al. IGF-1 deficiency impairs cerebral myogenic autoregulation in hypertensive mice. J Cereb Blood Flow Metab 2014; 34: 1887–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dunn KM, Renic M, Flasch AK, et al. Elevated production of 20-HETE in the cerebral vasculature contributes to severity of ischemic stroke and oxidative stress in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2008; 295: H2455–H2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Fan F, Geurts AM, Murphy SR, et al. Impaired myogenic response and autoregulation of cerebral blood flow is rescued in CYP4a1 transgenic dahl salt-sensitive rat. Am J Physiol Regul Integr Comp Physiol 2015; 308: R379–R390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Alonso-Galicia M, Drummond HA, K, Reddy K, et al. Inhibition of 20-HETE production contributes to the vascular responses to nitric oxide. Hypertension 1997; 29: 320–325. [DOI] [PubMed] [Google Scholar]
- 104.Sobey CG, Faraci FM. Effects of a novel inhibitor of guanylyl cyclase on dilator responses of mouse cerebral arterioles. Stroke 1997; 28: 837–843. [DOI] [PubMed] [Google Scholar]
- 105.Onoue H, Katusic ZS. The effect of 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) and charybdotoxin (CTX) on relaxations of isolated cerebral arteries to nitric oxide. Brain Res 1998; 785: 107–113. [DOI] [PubMed] [Google Scholar]
- 106.Feterik K, Smith L, Katusic ZS. Angiotensin-(1-7) causes endothelium-dependent relaxation in canine middle cerebral artery. Brain Res 2000; 873: 75–82. [DOI] [PubMed] [Google Scholar]
- 107.Yang G, Iadecola C. Activation of cerebellar climbing fibers increases cerebellar blood flow: role of glutamate receptors, nitric oxide, and cGMP. Stroke 1998; 29: 499–508. [DOI] [PubMed] [Google Scholar]
- 108.Zhang W, Potrovita I, Tarabin V, et al. Neuronal activation of NF-κB contributes to cell death in cerebral ischemia. J Cereb Blood Flow Metab 2005; 25: 30–40. [DOI] [PubMed] [Google Scholar]
- 109.Terai K, Matsuo A, McGeer EG, et al. Enhancement of immunoreactivity for NF-κB in human cerebral infarctions. Brain Res 1996; 739: 343–349. [DOI] [PubMed] [Google Scholar]
- 110.Gabriel C, Justicia C, Camins A, et al. Activation of nuclear factor-κ-B in the rat brain after transient focal ischemia. Molecular Brain Research 1999; 65: 61–69. [DOI] [PubMed] [Google Scholar]
- 111.Stephenson D, Yin T, Smalstig EB, et al. Transcription factor nuclear factor-kappa B is activated in neurons after focal cerebral ischemia. J Cereb Blood Flow Metab 2000; 20: 592–603. [DOI] [PubMed] [Google Scholar]
- 112.Leinhase I, Schmidt OI, Thurman JM, et al. Pharmacological complement inhibition at the C3 convertase level promotes neuronal survival, neuroprotective intracerebral gene expression, and neurological outcome after traumatic brain injury. Exp Neurol 2006; 199: 454–464. [DOI] [PubMed] [Google Scholar]
- 113.Burk AM, Martin M, Flierl MA, et al. Early complementopathy after multiple injuries in humans. Shock 2012; 37: 348–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bellander BM, Singhrao SK, Ohlsson M, et al. Complement activation in the human brain after traumatic head injury. J Neurotrauma 2001; 18: 1295–1311. [DOI] [PubMed] [Google Scholar]
- 115.Stahel PF, Flierl MA, Morgan BP, et al. Absence of the complement regulatory molecule CD59a leads to exacerbated neuropathology after traumatic brain injury in mice. J Neuroinflammation 2009; 6: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Fumagalli S, Perego C, Pischiutta F, et al. The ischemic environment drive s microglia and macrophage function. Front Neurol 2015; 6: 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation 2018; 15: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Färber K, Cheung G, Mitchell D, et al. C1q, the recognition subcomponent of the classical pathway of complement, drives microglial activation. J Neurosci Res 2009; 87: 644–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Orsini F, de Blasio D, Zangari R, et al. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci 2014; 8: 380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Stahel PF, Trentz O, Kossmann T, et al. Intrathecal levels of complement-derived soluble membrane attack complex (sc5b-9) correlate with blood-brain barrier dysfunction in patients with traumatic brain injury. J Neurotrauma 2001; 18: 773–781. [DOI] [PubMed] [Google Scholar]
- 121.Ricklin D, Hajishengallis G, Yang K, et al. Complement: a key system for immune surveillance and homeostasis. Nat Immunol 2010; 11: 785–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Leinhase I, Michael VM, Thurman JM, et al. Reduced neuronal cell death after experimental brain injury in mice lacking a functional alternative pathway of complement activation. BMC Neurosci 2006; 7: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Leinhase I, Rozanski M, Harhausen D, et al. Inhibition of the alternative complement activation pathway in traumatic brain injury by a monoclonal anti-factor B antibody: a randomized placebo-controlled study in mice. J Neuroinflammation 2007; 4: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kaczorowski SL, Schiding JK, Toth CA, et al. Effect of soluble complement receptor-1 on neutrophil accumulation after traumatic brain injury in rats. J Cereb Blood Flow Metab 1995; 15: 860–864. [DOI] [PubMed] [Google Scholar]
- 125.Longhi L, Perego C, Ortolano F, et al. C1-inhibitor attenuates neurobehavioral deficits and reduces contusion volume after controlled cortical impact brain injury in mice. Crit Care Med 2009; 37: 659–665. [DOI] [PubMed] [Google Scholar]
- 126.You Z, Yang J, Takahashi K, et al. Reduced tissue damage and improved recovery of motor function after traumatic brain injury in mice deficient in complement component C4. J Cereb Blood Flow Metab 2007; 27: 1954–1964. [DOI] [PubMed] [Google Scholar]
- 127.Yager PH, You Z, Qin T, et al. Mannose binding lectin gene deficiency increases susceptibility to traumatic brain injury in mice. J Cereb Blood Flow Metab 2008; 28: 1030–1039. [DOI] [PubMed] [Google Scholar]
- 128.Orsini F, Villa P, Parrella S, et al. Targeting mannose-binding lectin confers long-lasting protection with a surprisingly wide therapeutic window in cerebral ischemia. Circulation 2012; 126: 1484–1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Longhi L, Orsini F, de Blasio D, et al. Mannose-binding lectin is expressed after clinical and experimental traumatic brain injury and its deletion is protective. Crit Care Med 2014; 42: 1910–1918. [DOI] [PubMed] [Google Scholar]
- 130.Yu W, Le HW, Lu YG, et al. High levels of serum mannose-binding lectins are associated with the severity and clinical outcomes of severe traumatic brain injury. Clinica Chimica Acta 2015; 451: 111–116. [DOI] [PubMed] [Google Scholar]
- 131.Zangari R, Zanier ER, Torgano G, et al. Early ficolin-1 is a sensitive prognostic marker for functional outcome in ischemic stroke. J Neuroinflammation 2016; 13: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zanier ER, Zangari R, Munthe-Fog L, et al. Ficolin-3-mediated lectin complement pathway activation in patients with subarachnoid hemorrhage. Neurology 2014; 82: 126–134. [DOI] [PubMed] [Google Scholar]
- 133.de Blasio D, Fumagalli S, Orsini F, et al. Human brain trauma severity is associated with lectin complement pathway activation. J Cereb Blood Flow Metab 2019; 39: 794–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Osthoff M, Walder B, Delhumeau C, et al. Association of lectin pathway protein levels and genetic variants early after injury with outcomes after severe traumatic brain injury: a prospective cohort study. J Neurotrauma 2017; 34: 2560–2566. [DOI] [PubMed] [Google Scholar]
- 135.Helmy A, Carpenter KLH, Menon DK, et al. The cytokine response to human traumatic brain injury: Temporal profiles and evidence for cerebral parenchymal production. J Cereb Blood Flow Metab 2011; 31: 658–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Panerai RB. Assessment of cerebral pressure autoregulation in humans - A review of measurement methods. Physiol Meas 1998; 19: 305–338. [DOI] [PubMed] [Google Scholar]
- 137.Lassen NA, Christensen MS. Physiology of cerebral blood flow. Br J Anaesth 1976; 48: 719–734. [DOI] [PubMed] [Google Scholar]
- 138.Payne S. Cerebral flow in the brain control of blood autoregulation. Berlin: Springer, 2016, p. 125.
- 139.Ruesch A, Acharya D, Schmitt S, et al. Comparison of static and dynamic cerebral autoregulation under anesthesia influence in a controlled animal model. PLoS One 2021; 16: e0245291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Aaslid R, Lindegaard KF, Sorteberg W, et al. Cerebral autoregulation dynamics in humans. Stroke 1989; 20: 45–52. [DOI] [PubMed] [Google Scholar]
- 141.Tiecks FP, Lam AM, Matta BF, et al. Effects of the valsalva maneuver on cerebral circulation in healthy adults: a transcranial doppler study. Stroke 1995; 26: 1386–1392. [DOI] [PubMed] [Google Scholar]
- 142.Oblak JP, Zaletel M, Zvan B, et al. The effect of age on cerebrovascular reactivity to cold pressor test and head-up tilt. Acta Neurol Scand 2002; 106: 30–33. [DOI] [PubMed] [Google Scholar]
- 143.Czosnyka M, Pickard J, Whitehouse H, et al. The hyperaemic response to a transient reduction in cerebral perfusion pressure – a modelling study. Acta Neurochir 1992; 115: 90–97. [DOI] [PubMed] [Google Scholar]
- 144.van Beek AHEA, Claassen JAHR, Rikkert MGMO, et al. Cerebral autoregulation: an overview of current concepts and methodology with special focus on the elderly. J Cereb Blood Flow Metab 2008; 28: 1071–1085. [DOI] [PubMed] [Google Scholar]
- 145.Panerai RB. Transcranial doppler for evaluation of cerebral autoregulation. Clin Auton Res 2009; 19: 197–211. [DOI] [PubMed] [Google Scholar]
- 146.Madsen PL, Secher NH. Near-infrared oximetry of the brain. Prog Neurobiol 1999; 58: 541–560. [DOI] [PubMed] [Google Scholar]
- 147.Tas J, Beqiri E, van Kaam RC, et al. Targeting Autoregulation-Guided cerebral perfusion pressure after traumatic brain injury (COGiTATE): a feasibility randomized controlled clinical trial. J Neurotrauma 2021; 38: 2790–2800. [DOI] [PubMed] [Google Scholar]
- 148.Reinhard M, Rutsch S, Hetzel A. Cerebral autoregulation in acute ischemic stroke. Persp Med 2012; 1: 194–197. [Google Scholar]
- 149.Jordan JD, Powers WJ. Cerebral autoregulation and acute ischemic stroke. Am J Hypertens 2012; 25: 946–950. [DOI] [PubMed] [Google Scholar]
- 150.Møller K, Skinhøj P, Knudsen GM, et al. Effect of short-term hyperventilation on cerebral blood flow autoregulation in patients with acute bacterial meningitis. Stroke 2000; 31: 1116–1122. [DOI] [PubMed] [Google Scholar]
- 151.Tureen JH, Dworkin RJ, Kennedy SL, et al. Loss of cerebrovascular autoregulation in experimental meningitis in rabbits. J Clin Invest 1990; 85: 577–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Reinhard M, Neunhoeffer F, Gerds TA, et al. Secondary decline of cerebral autoregulation is associated with worse outcome after intracerebral hemorrhage. Intensive Care Med 2010; 36: 264–271. [DOI] [PubMed] [Google Scholar]
- 153.Svedung Wettervik T, Fahlström M, Enblad P, et al. Cerebral pressure autoregulation in brain injury and disorders–a review on monitoring, management, and future directions. World Neurosurg 2022; 158: 118–131. [DOI] [PubMed] [Google Scholar]
- 154.Huang Z, Huang PL, Ma J, et al. Enlarged infarcts in endothelial nitric oxide synthase knockout mice are attenuated by nitro-L-arginine. J Cereb Blood Flow Metab 1996; 16: 981–987. [DOI] [PubMed] [Google Scholar]
- 155.Hariharan A, Jing Y, Collie ND, et al. Altered neurovascular coupling and brain arginine metabolism in endothelial nitric oxide synthase deficient mice. Nitric Oxide 2019; 87: 60–72. [DOI] [PubMed] [Google Scholar]
- 156.Toda N, Ayajiki K, Okamura T. Cerebral blood flow regulation by nitric oxide: recent advances. Pharmacol Rev 2009; 61: 62–97. [DOI] [PubMed] [Google Scholar]
- 157.Broos K, Feys HB, de Meyer SF, et al. Platelets at work in primary hemostasis. Blood Rev 2011; 25: 155–167. [DOI] [PubMed] [Google Scholar]
- 158.Akgören N, Fabricius M, Lauritzen M. Importance of nitric oxide for local increases of blood flow in rat cerebellar cortex during electrical stimulation. Proc Natl Acad Sci U S A 1994; 91: 5903–5907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Stefanovic B, Schwindt W, Hoehn M, et al. Functional uncoupling of hemodynamic from neuronal response by inhibition of neuronal nitric oxide synthase. J Cereb Blood Flow Metab 2007; 27: 741–754. [DOI] [PubMed] [Google Scholar]
- 160.Schuman EM, Madison DV. Nitric oxide and synaptic function. Annu Rev Neurosci 1994; 17: 153–183. [DOI] [PubMed] [Google Scholar]
- 161.Toda N, Okamura T. The pharmacology of nitric oxide in the peripheral nervous system of blood vessels. Pharmacol Rev 2003; 55: 271–324. [DOI] [PubMed] [Google Scholar]
- 162.Freire MAM, Guimarães JS, Leal WG, et al. Pain modulation by nitric oxide in the spinal cord. Front Neurosci 2009; 3: 175–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Nathan C. Inducible nitric oxide synthase: what difference does it make? J Clin Invest 1997; 100: 2417–2423. [DOI] [PMC free article] [PubMed] [Google Scholar]



