Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is a devastating cerebral event that kills or debilitates the majority of those afflicted. The blood that spills into the subarachnoid space stimulates profound cerebral artery vasoconstriction and consequently, cerebral ischemia. Thus, once the initial bleeding in SAH is appropriately managed, the clinical focus shifts to maintaining/improving cerebral perfusion. However, current therapeutic interventions largely fail to improve clinical outcome, because they do not effectively restore normal cerebral artery function. This review discusses emerging evidence that perturbed cerebrovascular “myogenic reactivity,” a crucial microvascular process that potently dictates cerebral perfusion, is the critical element underlying cerebral ischemia in SAH. In fact, the myogenic mechanism could be the reason why many therapeutic interventions, including “Triple H” therapy, fail to deliver benefit to patients. Understanding the molecular basis for myogenic reactivity changes in SAH holds the key to develop more effective therapeutic interventions; indeed, promising recent advancements fuel optimism that vascular dysfunction in SAH can be corrected to improve outcome.
Keywords: Cerebral blood flow, cystic fibrosis transmembrane conductance regulator (CFTR), microvascular dysfunction, myogenic vasoconstriction, tumor necrosis factor
For neurologists tasked with saving patients suffering from subarachnoid hemorrhage (SAH), this review provides a mechanistic overview of the next frontier of therapeutic interventions. The insight provided calls attention to emerging evidence that perturbed cerebrovascular “myogenic reactivity” is a critical element underlying “delayed cerebral ischemia” (DCI) in SAH. Myogenic reactivity is a crucial microvascular process that potently dictates cerebral perfusion: indeed, this mechanism can explain why many current therapeutic interventions fail to improve outcome (e.g., triple H therapy) and is key to developing more targeted and successful interventions. As a caveat, this review's focus on myogenic mechanisms does not diminish the relevance of other pathological processes (e.g., blood barrier disruption, microthrombosis and cortical spreading) in the SAH injury process.1 In fact, we speculate that the ischemia caused by perturbed myogenic reactivity either causes or exacerbates these complications.
A brief clinical overview of subarachnoid hemorrhage
SAH is a devastating cerebral event that kills or debilitates the majority of those affected.2,3 Although SAH accounts for only 5% of all strokes (∼10 in 100,000 persons per year), the young age of incidence and poor prognosis instills a substantial societal burden: SAH accounts for 27% of all stroke-related years lost before the age of 65 (a measure of premature mortality) and therefore, closely rivals the most frequent stroke subtype, ischemic stroke, in terms of productive life years lost (38% for ischemic stroke).4,5 SAH carries an extremely high fatality rate (about 50%) and approximately 30–50% of the patients who survive are seriously debilitated.2,3 As these grim statistics suggest, current medical interventions display limited efficacy, and thus, SAH remains broadly regarded as a “large unmet medical need”.
Almost 85% of spontaneous SAH cases (i.e. cases not due to trauma) are caused by a ruptured cerebral aneurysm.5 In its initial phase, SAH is a medical emergency: bleeding into the subarachnoid space sharply elevates intracranial pressure, which ultimately causes cerebro-circulatory arrest if it is not swiftly relieved. Consequently, almost half of SAH patients die before reaching hospital.5 However, much of the treatable mortality and morbidity in SAH is caused by a pronounced secondary ischemic event, aptly termed “delayed cerebral ischemia or DCI”, that emerges 4-12 days after the initial hemorrhage.6,7 Thus, once the initial bleeding in SAH is appropriately managed, the clinical focus shifts to maintaining cerebral perfusion.
The therapeutic failure of targeting angiographic vasospasm
Delayed cerebral ischemia in SAH has been traditionally attributed to “angiographic vasospasm,” a radiographically identifiable vasoconstriction of large/proximal cerebral arteries: vasospasm is a frequent complication in SAH, it clearly associates with poor clinical outcome, and it is an intuitive cause of deficient cerebral perfusion.2,8 Clinical studies implicated endothelin 1 (ET-1), a potent endothelial-derived vasoconstrictor,9,10 as a causative factor by demonstrating a strong correlation between plasma/cerebrospinal fluid ET-1 levels and angiographic vasospasm severity in SAH patients.11–13 Experimental studies that targeting ETA, the “vasoconstrictive” ET-1 receptor subtype, demonstrated a clear benefit in terms of mitigating angiographic vasospasm:14: these results fueled numerous clinical trials, most notably CONSCIOUS.15–17 Unfortunately, while these clinical trials (that to date, include over 2600 SAH patients) demonstrate that ETA antagonism (clazosentan) successfully curtails angiographic vasospasm in SAH, the intervention does not improve clinical outcome.16,18
In retrospect, the decision to initiate large randomized trials for ET-1 receptor antagonists was not supported by robust experimental data on clinically relevant outcomes.14 Although angiographic vasospasm is a well-established predictor of cerebral ischemia/infarction in SAH,19–21 its sensitivity and specificity (both approximately 70%) are far from optimal.19 A meta-analysis of 55 experimental studies determined that the majority failed to assess whether ET-1 receptor antagonism changed cerebral blood flow (CBF) in SAH (only 3 of the 27 studies measured CBF); none of the studies investigated neurofunctional outcomes.14
Unfortunately, clinical trials are frequently initiated on limited, incomplete, and weak preclinical data. This longstanding issue has been recurrently addressed by the publication of scientific guidelines, with notable contributions from the Stroke Therapy Academic Industry Roundtable (STAIR)22,23 and Sena et al.24 In general, ideal interventions should (i) demonstrate efficacy in at least two species and in at least two laboratories that use different models; (ii) involve adequate physiological monitoring, including CBF measurements; and (iii) assess multiple functional and outcome measures, including brain injury volume, and neurological function. Individually, studies should adhere to good scientific practices by utilizing proper sample sizes, randomization, allocation concealment, blinding and case fatality reporting.22–24 In this light, the CONSCIOUS trial failure would almost certainly have been averted had the available data been scrutinized against these guidelines: critical measures were clearly missing and the studies were generally weak according to STAIR criteria.14 It is worrisome that scientific quality remains an underappreciated issue among those who conduct preclinical research,14,25 since there is no doubt that weak studies frequently overestimate drug efficacy.25 Thus, as the SAH field presses forward in its search for new therapeutic interventions, a concerted effort to assess all relevant clinical endpoints (e.g. CBF, neuronal injury, etc.) with rigorous scientific methodology is imperative.
Although ET-1 antagonists failed in clinical trials, two crucial conclusions can be formulated: first and foremost, while angiographic vasospasm is an informative parameter that correlates with disease severity and predicts clinical outcome, it is itself not the central determinant of ischemic injury.14,26–28 Indeed, there is ample evidence that SAH reduces CBF in the absence of angiographic vasospasm,29–34 indicative of a significant dissociation between these two endpoints. Second, distinct mechanisms drive constriction in the proximal and distal regions of the cerebrovascular tree; consequently, angiographic vasospasm is an insufficient endpoint for assessing new therapeutic interventions. Ultimately, the failure of the CONSCIOUS clinical trials compels a shift in attention from large cerebral arteries (i.e. angiographic vasospasm) to the cerebral microcirculation. In fact, augmenting small resistance artery vasoconstriction35 would compromise one of the most significant determinants of cerebral perfusion: CBF autoregulation.
Cerebral blood flow autoregulation
Without question, minimum perfusion levels must be maintained in the brain, because neurons have high metabolic demand and minimal energy reserves. Large perfusion reductions rapidly compromise neuronal function and lead to infarction; however, even in the absence of “symptomatic hypoperfusion” (i.e. reductions in blood flow that cause dizziness or other overt symptoms), inadequate cerebral perfusion slowly compromises neuronal function and drives cognitive decline.36 While hypoperfusion has obvious negative consequences, the brain is also detrimentally impacted by hyperperfusion (appropriately termed “cerebral hyperperfusion syndrome”).37 Specifically, insufficient vascular resistance (i) promotes hydrostatic edema formation, due to increased Starling forces37 and (ii) can cause pressure-induced damage of the fragile capillary networks.38,39 Cerebral perfusion, therefore, must be tightly regulated with an exquisite level of precision.
Cerebral blood flow autoregulation refers to the intrinsic ability of the distal cerebral resistance arteries to match their flow resistance to the prevalent perfusion pressure, thereby maintaining constant perfusion over a wide range of systemic pressures.40 Although the precise pressure range governed by autoregulation is subject to some dispute,41 the widely accepted autoregulatory range in humans is 60-150 mmHg. Technically, the term autoregulation refers only to the maintenance of constant flow in response to changes in pressure; this process is achieved by a mechanism localized to resistance arteries (named “the myogenic response”). When neuronal metabolism necessitates an increase in perfusion, “metabolic” and “neurovascular” coupling mechanisms superimpose on autoregulation, to locally match perfusion to neuronal energy demand. As examples of the latter mechanisms, carbon dioxide tension, adenosine (a metabolic product), nitric oxide and sympathetic activity all locally influence vascular tone and hence, regional perfusion within the brain.42,43 Separating local influences from autoregulation in vivo is challenging: consequently, our understanding of how disease processes alter autoregulation is rather rudimentary. Non-invasive imaging studies clearly demonstrate that SAH compromises autoregulation,44–46 but the mechanistic basis requires an experimental system that characterizes a resistance artery's myogenic response (i.e. its pressure/diameter relationship) in vitro to eliminate the confounding influences of local metabolic and neurovascular mechanisms.
The myogenic response
Over a century ago, Sir William Bayliss made the seminal observation “when now the pressure was raised inside the artery it was seen at first to swell, but immediately, and while the mercury was still kept at its height, a powerful contraction took place.”47 This observation, named the myogenic response,48–56 describes the dynamic adjustment of blood vessel diameter to changes in perfusion pressure that forms the basis of local blood flow autoregulation. Specifically, the myogenic response is vasoconstriction in response to an increase in transmural pressure (i.e. the pressure across the vessel wall) or vasodilation in response to a decrease in transmural pressure.
As the etymology of its name implies (myo = muscle, genic = generated), the myogenic response originates from the vascular wall smooth muscle layer.57–59 Vascular smooth muscle cells are continuously exposed to changes in mechanical load (i.e. circumferential stretch), due to changes in blood pressure: the smooth muscle cells respond to this stimulus by actively adjusting their contractile force and ultimately, the level of active constriction exhibited by a artery. Notably, a vessel will constrict to a diameter smaller than its initial diameter in response to an increase in transmural pressure,60,61 indicating that the controlled variable in myogenic signaling is not vessel diameter per se. Instead, in accordance with the law of Laplace (wall tension = transmural pressure gradient × vessel radius), myogenic vasoconstriction endeavors to normalize pressure-induced increases in vessel wall tension, a concept supported by a direct correlation between changes in vessel wall tension and intracellular myogenic vasoconstriction signaling events.60,62,63
Not all arteries within the vascular tree display myogenic reactivity: large conduit arteries, which are tasked with rapidly conducting large volumes of blood, do not display pressure-stimulated vasoconstriction.64 Instead, it is the small, muscular resistance arteries (30–250 µm in diameter), located just proximal to the capillary beds, that actively control blood flow in response to pressure and integrate the modulatory influences of tissue-derived signaling molecules.65,66 Even here, resistance arteries display a gradient of myogenic reactivity that is inversely related to luminal diameter. For example, in the hamster cheek pouch, third-order resistance arteries (28 µm) exhibit ∼23% greater myogenic index (i.e., the slope of the pressure-diameter relationship) than the first-order arteries (81 µm).67 This indicates that the smaller resistance arteries are of particular functional importance for blood pressure regulation,68,69 the protection of downstream capillary integrity38,39 and minimization of edema formation.70
The molecular basis for the myogenic response has been extensively reviewed;52,53,55,56 since this is a substantial topic in its own right, this review will provide a simplified and abbreviated overview. The molecular signaling pathways that control the myogenic response involve multiple signaling streams, broadly separated into: (i) mechanosensor components that register the pressure/stretch stimulus and transduce physical forces into intracellular signals, (ii) the activation of calcium release and calcium sensitization mechanisms, and (iii) the convergence of these mechanisms to processes that phosphorylate the myosin light chain and initiate cross-bridge cycling.
No single mechanosensor has been established as the master regulator of myogenic reactivity: instead, there appears to be a diverse array of putative mechanosensors, including stretch-activated ion channels (e.g. transient receptor potential nonselective cation channels71–73), G-protein coupled receptors (e.g. angiotensin type I receptor,74 integrins (e.g. α5β1 and αvβ3 heterodimers75,76) and most recently, membrane-bound tumor necrosis factor.77 These mechanosensitive elements are redundantly organized, which likely preserves a fully functional myogenic mechanism in several settings.
Mechanosensor activation increases intracellular calcium levels and stimulates signaling processes that amplify responsiveness to calcium, termed calcium sensitization.52,78 Initially, vascular smooth muscle cells depolarize in response to increased wall tension,79–81 which stimulates calcium influx through the dihydropyridine-sensitive L-type voltage gated calcium channels;81 calcium release from the sarcoplasmic stores also occurs.82 Cytosolic calcium binds to calmodulin, leading to myosin light chain kinase (MLCK) activation and subsequent myosin light chain 20 (MLC20) phosphorylation.55 MLC20 phosphorylation is the critical signaling event in pressure- and agonist-induced vasoconstriction62,83,84 that activates actin-myosin filament interaction and gliding.55 Concurrent with these calcium-dependent mechanisms, calcium sensitization processes increase the level of MLC20 phosphorylation for a given concentration of cytosolic calcium:52 this is ultimately achieved by inhibiting the activity of myosin light chain phosphatase (MLCP).55 There are many signaling mechanisms meditating calcium sensitization: notable examples include Rho kinase and protein kinase C signaling.55,78,83,85–87
Subarachnoid hemorrhage augments myogenic reactivity
Surprisingly, despite its fundamental role in CBF autoregulation,40,88 only a handful of investigations have assessed myogenic reactivity in SAH. Although few, these studies clearly demonstrate that SAH augments cerebral artery myogenic tone in mouse,35 rat,89,90 rabbit91,92 and canine93,94 models. Intuitively, augmented myogenic reactivity must reduce CBF; however, the mechanistic basis for these augmented responses and its relationship to critical in vivo parameters (e.g. CBF and neuronal injury) have not been extensively explored.
In mouse olfactory cerebral resistance arteries, SAH augments myogenic tone in a time-dependent manner: the enhancement is maximal at two days post-SAH induction; it then declines (but does not fully resolve) by five days post-SAH induction.35 Remarkably, this time course is consistent with vasospasm severity reported by Lin and coworkers95 in a similar mouse model. Myogenic tone does not increase when saline is injected into the intracranial space, confirming that the augmented myogenic tone is specifically caused by the presence of blood in the intracranial space. The preservation of a normal phenylephrine dose–response relationship indicates that the augmentation is specific to myogenic mechanisms, rather than an increase in general contractility.35
Assuming that the olfactory artery reasonably replicates effects within the distal microcirculation, we can predict the effect on CBF by using Poiseuille's law. To review Poiseuille's law: F = π ΔP r4 / 8 η L, where F is the flow, ΔP is the pressure differential, r is the vessel radius, η is the fluid viscosity, and L is the length. Using the myogenic tone measures collected by Yagi and coworkers (the calculations here use the myogenic tone measured at 60 mmHg in Figure 1A from Yagi and coworkers),35 we calculate the flow effect as follows, defining the maximum possible flow as “1” (i.e., the flow at maximum diameter of “1”):
(i) Flow varies with r4;
(ii) the flow rate with 13% myogenic tone is (0.87)4 =0.573 (57% of maximum);
(iii) the flow rate with 29% myogenic tone is (0.71)4 =0.254 (25% of maximum);
(iv) the flow reduction is (0.573-0.254)/0.573 × 100 =55.7%.
Figure 1.
The myogenic mechanism underlies the failure of current therapeutic interventions. Shown are schematic tracings of myogenic responses and cerebral autoregulation, as would be observed in the cerebral microcirculation prior to and following subarachnoid hemorrhage (SAH). The myogenic response adjusts vascular resistance proportionally to pressure, resulting in constant flow over a wide range of perfusion pressures. In SAH, myogenic reactivity is augmented: autoregulation persists, but perfusion is reduced. The reduced perfusion in SAH results in secondary ischemic injury. Therapeutic intervention with vasodilators overrides the myogenic response, which compromises autoregulation. Perfusion increases, but pressure is transmitted to the delicate microcirculation, resulting in microvascular injury and edema formation. Therapeutic intervention with “Triple H Therapy” (hypertension, hypervolemia and hemodilution) confers minimal benefit since: (i) within the autoregulatory range, the myogenic response counteracts the pressure increase, resulting in no flow change; or (ii) beyond the autoregulatory range (“breakthrough” pressures), pressure is transmitted to the delicate microcirculation, resulting in microvascular injury and edema formation. Restoring normal myogenic reactivity is the most effective means to improve perfusion without secondary injury.
As predicted by the myogenic data, Yagi and coworkers observe that forebrain perfusion is, in fact, reduced by 56% in SAH 35. In addition to entirely explaining the CBF reduction, this calculation illustrates that relatively small changes in myogenic tone (i.e. from 13% to 29%) can cause serious perfusion impairment.
Setting perfusion thresholds that faithfully predict neuronal injury is extremely challenging. Ischemia is not defined by the single parameter of blood flow: rather, it represents the balance between blood supply and metabolic rate. Setting a singular threshold, therefore, ignores regional variation in metabolic need; by association, estimating a threshold from a single region does the same. Adding to this complexity, ischemic susceptibility varies across different species and even within different mouse strains.96,97 Defining a perfusion threshold for injury, therefore, is at best, an estimate.
Nevertheless, several experimental and clinical investigations have attempted to deduce a perfusion threshold, by assessing CBF in ischemic tissue (i.e. most often, the penumbra region of a focal infarct) and then discriminating the tissue that progresses to infarct (including the ischemic core) from the tissue that recovers. As a reasonable estimate, reducing CBF by >40% significantly risks permanent neuronal injury, with the severity proportional to both the degree and duration of hypoperfusion.98–101 Thus, the 56% CBF reduction measured by Yagi and coworkers is very likely to cause the neuronal apoptosis/degeneration and reduced neurofunctional scores observed at two days post-SAH induction.35
In order to therapeutically correct exaggerated myogenic vasoconstriction in SAH, a significant investigative investment is required in order to understand the molecular basis for altered responses. However, several factors discourage this investment: the molecular mechanisms that control myogenic signaling are extraordinarily complex and display significant heterogeneity across different species and vascular beds;56 the pressure myography assessment method is slow and labor intensive; and a high level of training is required to develop the necessary microsurgical skills. In the absence of the key mechanistic insight into how myogenic responsiveness changes in SAH, the current therapies utilized are generally ineffective and, in certain instances, are counterproductive.
Current therapeutic interventions in SAH
Impaired cerebral autoregulation is recognized as a strong independent predictor of both delayed cerebral ischemia44 and negative outcome45,46 in SAH. This is not surprising, given that blood flow autoregulation is required to maintain the structural and functional integrity of the brain. However, correcting autoregulatory dysfunction necessitates the specific correction of perturbed myogenic mechanisms (Figure 1). In this regard, current therapeutic interventions are non-selective and attempt to improve cerebral perfusion by “forcibly increasing flow” (e.g. “Triple H” and hypertensive therapy), physically imposing dilation (e.g. angioplasty) or indiscriminately interfering with vasoconstrictor mechanisms (e.g. the calcium channel antagonist nimodipine) 102.
Triple-H Therapy, classically defined as hypertension, hypervolemia, and hemodilution, is intended to improve cerebral hemodynamic flow by increasing perfusion pressure, expanding intravascular volume, and decreasing blood viscosity.103,104 The rationale behind Triple H and hypertensive therapies is an assumption that cerebral vasospasm eliminates cerebral blood flow autoregulation.103,104 In this scenario, if the vasospasm cannot be therapeutically targeted, the only variables in Poiseuille's law that can be manipulated to improve perfusion are pressure (ΔP) and viscosity (η). However, the clear flaw in this rationale is that autoregulation, in fact, persists in SAH (i.e. increasing pressure stimulates vasoconstriction), with a shifted autoregulatory window that actively maintains CBF at an insufficient level.46,105,106 Thus, Triple-H therapy can be expected to increase CBF only at pressures higher than the autoregulatory window, a maneuver that risks microvascular damage to all microcirculatory beds exposed to systemic pressures.38,39 This presumably explains why Triple H therapy frequently fails to improve CBF or outcome in SAH (Figure 1).107–109 Despite the clear lack of objective evidence from controlled clinical trials that Triple-H therapy or any its separate components increase CBF,109 current clinical guidelines110 recommend treating delayed ischemia in SAH with hypertension, based primarily on anecdotal observations110 and individual case reports.111,112 The methods and manner of administrating Triple-H therapy are highly variable113 and the therapy can cause serious medical complications, including cardiopulmonary failure, renal dysfunction, exacerbation of cerebral edema, and the potential rupture of unsecured intracranial aneurysms.27,102,104 There is no consensus on how to administer the therapy, nor is there agreement on clinical endpoints that should guide the therapy.113
Endovascular therapy (e.g. transluminal balloon angioplasty) may be employed prophylactically or as a “rescue procedure”, if severe angiographic vasospasm is quickly detected.114 The therapeutic window is narrow115 and the prospects of successful treatment are likely limited to the most severe cases, where proximal artery vasoconstriction reaches near-occlusive levels. For obvious reasons, endovascular therapy will not alleviate ischemia caused by distal cerebrovascular constriction: this may explain why prophylactic angioplasty reduces vasospasm, but generally fails to improve clinical outcome.116 The evidence supporting angioplasty is sparse and no large, random controlled trials have been conducted.
Several Calcium Channel Blockers have been investigated as a therapeutic intervention in SAH, with nimodipine and nicardipine receiving the majority of attention.27,102 Nimodipine, a dihydropyridine that blocks L-type calcium channels, has purported selectivity for cerebral vascular smooth muscle and is the only medication approved by the FDA for treating “symptomatic vasospasm” (i.e. the symptoms of delayed cerebral ischemia in SAH). Orally administered nimodipine clearly improves clinical outcome;27,102,117–119 however, experimental120 and clinical117,118,121,122 evidence indicate that the neurological improvement is not associated with a reversal of angiographic vasospasm or the normalization of CBF: the improved outcomes, therefore, are attributed to direct neuroprotection.123–126 Indeed, neurons express higher levels of L-type Ca2+ channels than vascular cells127 and topical, intra-arterial nimodipine application at high doses appears to be the only means of reliably inducing vasodilation.128–130 Interestingly, antagonizing voltage-dependent calcium channels with magnesium fails to improve clinical outcome131,132 and clinical outcome results for nicardipine, an alternative dihydropyridine that blocks L-type calcium channels, have been mixed.133,134
The search for the mechanism responsible for augmented myogenic reactivity in subarachnoid hemorrhage
There is general agreement that the breakdown of blood in the subarachnoid space causes the delayed cerebral artery vasoconstriction and ischemia.135 Several classic in vitro studies involving oxyhemoglobin and erythrocytes136–138 strongly support this premise, as does clinical evidence that delayed cerebral ischemia and vasospasm correlate with the volume of blood in the subarachnoid space.139,140 However, there are many ways that subarachnoid blood can elicit cerebral artery vasoconstriction: the most notable hypotheses involve nitric oxide scavenging,141,142 oxidant stress/free radical generation,141,143 the induction of inflammatory responses,141,144 and the stimulation of vasoconstrictor release (e.g. endothelin-1).145 These proposed mechanisms are not mutually exclusive and they themselves elicit a myriad of pleiotropic effects. Thus, the task of selecting a single therapeutic target that will ameliorate ischemic injury in SAH is difficult, as exemplified by the dismal success rate of most new interventions in clinical trials.146
As stated earlier, the most significant factor hindering the development of more precise therapeutic strategies to correct/prevent microvascular constriction is a distinct lack of mechanistic knowledge of how cerebral resistance artery reactivity changes in SAH. In fact, even within the limited number of reports utilizing isolated artery models, many of these investigations do not explore myogenic mechanisms per se (i.e. they do not directly address how SAH augments myogenic tone91 or they study biomechanical/histological changes93,94). Nevertheless, several propositions, including endothelial dysfunction, reduced potassium channel activity, and increased 20-HETE signaling, are sound hypotheses: these mechanisms clearly modulate or drive myogenic reactivity and they appear to be profoundly perturbed in the pathological setting. Yet, several significant issues have hindered their development into therapeutic interventions: (i) although there are clear indications that SAH perturbs microvascular function,147,148 mechanisms promoting angiographic vasospasm (i.e. large artery vasoconstriction) have received an unbalanced share of clinical attention, as classic investigations149,150 concluded that vasospasm was the primary culprit responsible for DCI; (ii) there appears to be limited availability of clinically viable therapeutic interventions that modulate these pathways; and (iii) tested interventions have yet to be comprehensively studied with CBF and neuronal injury endpoints. To elaborate further, the next three subsections will briefly review the status of each of these proposed mechanisms.
Endothelial dysfunction in subarachnoid hemorrhage
SAH is known to profoundly alter endothelial function in cerebral arteries.1 Since the endothelium constitutively releases vasodilating factors (e.g. endothelial derived relaxing and hyperpolarizing factors; EDRFs and EDHFs), it is reasonable to speculate that endothelial dysfunction augments myogenic reactivity. In this regard, there is ample evidence that SAH impairs endothelial-dependent vasodilator responses in both experimental151–154 and clinical155–157 settings. Whether these impaired responses are truly endothelial in origin, however, is controversial. While certain studies link attenuated endothelial responses to impaired vasodilator release,158,159 numerous others show that vasodilator deficits are, in fact, an inability of smooth muscle cells to transduce the endothelial signals.151–155These latter studies prompted two small clinical trials testing sildenafil, a cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase type 5 (PDE5) inhibitor that prolongs endothelial-derived signals transduced by smooth muscle cells: their results suggest that sildenafil may alleviate angiographic vasospasm,160 but does not improve brain perfusion.161
With respect to myogenic reactivity, demonstrating that SAH alters vasodilator responsiveness is not, in and of itself, sufficient to implicate endothelial dysfunction as a causal mechanism augmenting myogenic vasoconstriction. To our knowledge, only one isolated resistance artery investigation has directly assessed the endothelial contribution to augmented myogenic reactivity in SAH.89 In this study, SAH induced a myogenic tone augmentation that persisted following endothelial denudation, clearly demonstrating the existence of a smooth muscle cell mechanism. Since the degree of augmentation (i.e. the absolute tone difference between control and SAH) did not markedly diminish with denudation, this smooth muscle cell mechanism appears to be the predominant mechanism enhancing myogenic tone.89 A second isolated resistance artery investigation has since provided indirect corroborating evidence, based on the preservation of phenylephrine dose-response sensitivity in SAH.35 Although no direct assessments of endothelial contributions were made, the authors precluded an endothelial mechanism on the basis that compromised endothelial vasodilatory influences should similarly affect both myogenic reactivity and vasoconstrictor responses.35 In fact, the preservation of “general contractility” observed in both studies (to potassium and phenylephrine, respectively)35,89 points to a specific effect on the myogenic mechanism. Consistent with this conclusion, “unpressurized” (5-10 mmHg) cerebral arteries from sham and SAH have similar membrane potentials and intracellular calcium levels,89 while pressure-stimulated depolarization and calcium elevations are markedly enhanced in arterioles from SAH mice, relative to sham controls.89,92
Membrane potential and potassium channels in subarachnoid hemorrhage
Depolarization is a critical initiator of the myogenic response that activates voltage-sensitive calcium channels and leads to extracellular calcium influx. In this regard, potassium channels are prominent regulators of vascular tone, as only a handful of channels need to open in order to achieve meaningful changes in membrane potential.162,163 Opening potassium channels increases potassium conductance, causing potassium efflux and hyperpolarization (until the −85 mV equilibrium potential for potassium is reached). In cerebral arteries, voltage gated potassium channels (Kv) and large conductance calcium-activated potassium channels (BK) dominate potassium conductance in smooth muscle cells and play a key role in limiting depolarization.163
Perturbed membrane potential regulation was one of the earliest hypotheses advanced to explain underlying basis of vascular constriction in SAH. Indeed, enhanced smooth muscle membrane potential depolarization clearly contributes to the augmented arteriolar tone observed in SAH.89,90,92,164,165 Several studies demonstrate that SAH reduces potassium currents in cerebral artery smooth muscle cells that, as expected, correlate with depolarization and enhanced constriction.90,164,165 Experiments using oxyhemoglobin to mimic the effect of the subarachnoid blood provide solid corroborating evidence.166–168 The precise channels affected, however, are less clear: Kv1.5, Kv2.1 and Kv2.2 transcripts and protein levels are reduced in SAH,169 although functional measures suggest that Kv2 and Kv7 family members are the pertinent functional targets.164,170 The involvement of BK channels is contradictory: Koide and co-workers showed that SAH causes a distinct reduction in calcium spark-induced transient BK currents,92 while Jahromi and co-workers found that virtually all BK channel characteristics are unaffected in SAH.171
To our knowledge, Kv7 channels are the only Kv channels that have been therapeutically targeted: Retigabine or Celecoxib both attenuate angiographic vasospasm in experimental SAH.170 Neurological outcomes were not assessed in this study170; however, both therapeutic entities are utilized clinically and Kv7 channels are known to regulate cerebral artery myogenic responses.172 Thus, it will be intriguing to follow further developments in this field. One caveat to consider, however, is that potassium channel modulating therapeutics are unlikely to specifically target the pathological mechanism augmenting myogenic tone: these therapeutics will possess broad vasodilatory properties, and therefore likely suffer similar pitfalls as other general vasodilators. This may explain why calcitonin-gene-related peptide, a putative ATP-dependent potassium channel activator,173 shows experimental promise,174 but has yet to demonstrate a clear clinical benefit in small preliminary trials.175,176
20-HETE in subarachnoid hemorrhage
Arachidonic acid metabolites are well-established modulators of vascular tone.177 The cytochrome P450 metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) rose to prominence as a vasoconstrictor following the revelation that it plays a key role in hypertension.178 Within the vascular wall, 20-HETE is synthesized predominantly by smooth muscle cells.178 Many vascular beds, including the cerebral microcirculation,179 utilize 20-HETE as a signaling molecule in smooth muscle contractility, proliferation, angiogenesis and inflammation.178,180 In cerebral arteries, 20-HETE potently stimulates vasoconstriction by promoting depolarization, calcium influx and the activation of calcium sensitization mechanisms, the latter of which notably involve protein kinase C (PKC) and Rho kinase activation.178,181,182 These vasoconstrictor mechanisms clearly overlap with known myogenic mechanisms, and thus, it is not surprising that 20-HETE is a prominent regulator of cerebral artery myogenic responsiveness.181,183,184
Observations that SAH stimulates fatty acid release185–187 fuelled speculation that 20-HETE could be a relevant metabolite that drives vasospasm and ischemic injury. In accordance with this hypothesis, cerebrospinal 20-HETE levels are elevated in SAH patients.188–190 However, its predictive value for putative vascular endpoints is rather poor. Specifically, while cerebrospinal 20-HETE clearly associates with neurological deterioration, its relationship to angiographic vasospasm, delayed cerebral ischemia, and infarction is weak.189,190 Siler and co-workers observed that elevated cerebrospinal 20-HETE levels measured within the first 96 h of admission predicts delayed cerebral ischemia; however, 20-HETE levels fall and are no longer predictive at the time when the ischemia actually occurs.191 While cerebrospinal 20-HETE levels may be an appropriate biomarker to identify patients at high risk for unfavorable neurological outcomes, it remains unclear whether the 20-HETE profiles for cerebrospinal fluid and the vascular wall align. This could explain the discrepancy between these clinical results that, at best, weakly correlate 20-HETE to DCI and the strong experimental evidence demonstrating that 20-HETE synthesis blockade improves vasospasm and CBF reductions in SAH.184,192–194 Currently, there is paucity in experimental studies examining 20-HETE as a therapeutic target in SAH: directly assessing whether 20-HETE augments myogenic reactivity in SAH, therefore, represents a key priority.
Tumor necrosis factor augments myogenic reactivity in subarachnoid hemorrhage
Recently, Yagi and co-workers have made significant advancements in our molecular understanding for how myogenic reactivity changes in SAH.35 Their study built on mechanistic insight gained from an etiologically distinct model of heart failure, where tumor necrosis factor (TNF) augments cerebral artery myogenic tone and disrupts CBF autoregulation.195,196 Briefly, previous work by this group demonstrates that myocardial infarction induces pathological vascular smooth muscle cell TNF expression,196 which down-regulates the cystic fibrosis transmembrane conductance regulator (CFTR) by an autocrine/paracrine mechanism.195 As a critical transporter involved in smooth muscle cell sphingosine-1-phosphate (S1P) degradation,195 CFTR down-regulation enhances S1P signaling and pro-constrictive effects in the context of myogenic vasoconstriction.195,196 A composite of this signaling mechanism, which has been constructed over decades,35,39,52,56,85,195–200 is presented in Figure 2.
Figure 2.
Current and proposed molecular mechanisms augmenting myogenic reactivity in subarachnoid hemorrhage. A mechanosensitive complex localized to the plasma membrane initiates the myogenic response, resulting in membrane potential depolarization, calcium entry and extracellular signal-regulated kinase 1 and 2 (ERK1/2) activation. ERK1/2 phosphorylates sphingosine kinase 1 (Sphk1), eliciting its translocation to the plasma membrane and synthesis of sphingosine-1-phosphate (S1P) from sphingosine. Intracellular S1P modulates pressure-stimulated calcium elevation, which activates calmodulin (CAM), myosin light chain kinase (MLCK) and the contraction apparatus. Extracellular S1P activates the S1P2 receptor subtype (S1P2R) to activate the RhoA/Rho kinase (ROCK) signaling pathway, which inhibits myosin light chain phosphatase (MLCP). Inhibiting MLCP enhances MLCK's activation of the contractile apparatus. Extracellular S1P is sequestered from S1P2R by the cystic fibrosis transmembrane conductance regulator (CFTR), which transports S1P across the plasma membrane for dephosphorylation by S1P phosphohydrolase 1 (SPP1). In subarachnoid hemorrhage (SAH), the induction of inflammatory tumor necrosis factor (TNF) signaling curtails CFTR gene expression (not shown) and accelerates CFTR degradation mechanisms: this results in higher S1P bioavailability for S1P2R signaling and increases activation of the contractile apparatus. In experimental settings, preventing TNF signaling (etanercept; ETN) or antagonizing S1P2R signaling (JTE013) successfully blocks this pathological process. Therapeutics that increases CFTR expression (Lumacaftor) or activity (Ivacaftor) would be expected to have similar efficacy in terms of blocking the pathological enhancement of S1P/S1P2R signaling: their value as a therapeutic intervention awaits experimental validation.
As a framework for how cerebrovascular reactivity changes in SAH, this mechanism is attractive for two key reasons: first, inflammation undoubtedly occurs in SAH144 and thus it is conceivable that heart failure and SAH elicit the same functional effect on cerebral artery myogenic tone. Indeed, TNF levels peak in a timeframe consistent with delayed cerebral ischemia in SAH201,202 and associate with angiographic vasospasm, abnormal cerebral flow velocities, and poor clinical outcome.203–206 Second, S1P is a potent regulator of cerebral artery tone35,195,196,207–211 and prominently regulates and coordinates pressure-dependent calcium responses,56,85,197 putatively providing the upstream link to the pressure-stimulated depolarization and calcium elevations mentioned above.89,90,92,212
Consistent with the heart failure model, Yagi and co-workers observed that SAH induces robust vascular TNF expression in olfactory cerebral arteries, predominantly localized to the smooth muscle cells within the microvascular wall.35 Using distinct inhibitory strategies, including smooth muscle cell-targeted TNF gene deletion, Yagi and co-workers conclusively demonstrated that smooth muscle cell TNF expression drives the SAH-mediated enhancement of myogenic vasoconstriction.35 Therapeutically sequestering TNF (etanercept) eliminates the SAH-mediated myogenic tone augmentation, normalizes cerebral perfusion, reduces neuronal injury and prevents neurofunctional deficits.35 These latter observations have since been independently corroborated: etanercept reduces apoptosis and improves neurological scores in a rat model of SAH.213 Also consistent with the heart failure mechanism,195,196 antagonizing S1P receptors (JTE013) has the same therapeutic benefits as sequestering TNF: normalization of myogenic tone, attenuated neuronal injury, and the prevention of neurofunctional deficits.35 Again, recent reports independently corroborate these therapeutic observations: fingolimod, a functional S1P receptor antagonist,214 (i) preserves vascular reactivity and improves neurofunctional outcome in a rat SAH model215 and (ii) improves clinical outcome in a similar type of hemorrhagic stroke (supratentorial intracerebral hemorrhage).216
Two significant features of this work should not be overlooked. First, both therapeutic interventions (etanercept and JTE013) restricted their effect to the SAH-mediated enhancement myogenic tone:35 neither intervention reduced myogenic tone below the sham level; they had no effect in sham mice; nor did they affect phenylephrine responses, indicating that general contractility was not compromised. This specificity confers clear advantages over conventional therapies that non-specifically blunt vasoconstriction and/or disrupt autoregulation. Second, both interventions affected all endpoints similarly,35 strengthening the intuitive concept that myogenic tone, CBF, and neuronal injury are causally linked, rather than simple correlations.
The next frontier – CFTR as a vascular regulator
Although Yagi and co-workers conclude that anti-TNF therapy can be used to improve neurological outcome in SAH,35 the immune system suppression associated with the intervention may further threaten these critically ill patients by increasing their risk of infection.217 Similarly, S1P signaling is central to many cellular processes (e.g. endothelial barrier function218,219): S1P receptor antagonism,220 therefore, may carry a significant risk of secondary effects. Of the therapeutic targets their proposed mechanistic concept identifies (Figure 2),35,195 the cystic fibrosis transmembrane conductance regulator (CFTR) emerges as an attractive and logical choice for correcting myogenic reactivity within the cerebral microcirculation for three key reasons: (i) it is the proposed mechanistic basis for TNF's tone-enhancing effect,195 (ii) FDA-approved therapeutics that augment CFTR activity are available221,222 and (iii) CFTR therapeutics are not known to compromise immune function and likely have fewer secondary effects than S1P receptor antagonism.
CFTR is classically known for its critical role in lung epithelial chloride secretion;223 however, as an ATP-binding cassette (ABC) transporter that functions as an ion channel, it possesses the undeniably unique feature that it consumes energy mediating the passive diffusion of chloride ions.224 In many cells, the equilibrium potential for chloride is similar to the membrane potential: activating or inhibiting CFTR activity, therefore, does not induce significant chloride flux or change membrane potential. However, as reviewed by Bulley and Jaggar, smooth muscle cells possess high intracellular chloride levels, and the equilibrium potential for chloride is estimated to fall between -30 and -20 mV.225 Here, opening CFTR channels will result in chloride efflux, except when the smooth muscle cells are already highly depolarized. Although chloride efflux should have a depolarizing effect, changing membrane potential will influence other channels involved in membrane potential maintenance (e.g. inward rectifier and voltage dependent potassium channels). Thus, it is not straightforward to predict how changes in CFTR activity impact membrane potential. Since CFTR's unitary conductance is relatively low (∼10 pS),226 other ion channels (both cation and anion) likely dominate the control of membrane potential. This presumably permits CFTR's chloride independent functions (e.g. transporter activities) to have a more pronounced role in modulating vascular reactivity. In this regard, CFTR regulates the transport of several charged molecules,227–229 including S1P.195,230 Specifically, CFTR constitutively transports extracellular S1P across the plasma membrane and into the intracellular compartment.195 This process diverts extracellular S1P from interacting with cell surface receptors, forming a rapid and effective basis to regulate S1P receptor activity.195,230 Increasing CFTR-mediated S1P transport sequesters S1P from its pro-constrictive S1P2 receptors,195 which attenuates vasoconstriction
In both olfactory35 and posterior195 cerebral arteries, inhibiting CFTR activity (e.g. via gene deletion or inhibitory compounds) augments myogenic tone; this augmentation can be eliminated by S1P receptor antagonism (JTE013). These effects are consistent with CFTR's role in transporting/sequestering S1P,195 as opposed to changes in chloride efflux. The loss of CFTR function does not alter phenylephrine responses,35,195 indicating restricted effects on myogenic mechanisms, rather than effects on general contractility. Protein expression analyses confirm that SAH down-regulates CFTR protein expression by a TNF-dependent mechanism.35 The strong correlation between CFTR expression and cerebral artery myogenic tone35,195 drives the proposition that CFTR therapeutics may confer benefits in SAH treatment.
As a caveat, we must note that CFTR is also expressed in endothelial231,232 and neuronal233,234 cells, with functions relevant to glycocalyx swelling (endothelial),231 vesicular transport (neuronal)235 and neuronal excitability.236 The presence of CFTR in these cell types raises the prospect that CFTR therapeutics could confer vascular reactivity and/or neuroprotective benefits in addition to the smooth muscle cell localized effect on S1P signaling. Thus, should CFTR therapeutics prove beneficial in SAH, follow-up studies utilizing cell-specific gene deletion models will be required to mechanistically localize the site of action. An obvious initial choice would be a smooth muscle cell-specific gene deletion model, which successfully characterized smooth muscle cell TNF as the relevant TNF pool augmenting myogenic reactivity in SAH.35
Therapeutic strategies to restore deficient CFTR function
The incidence of cystic fibrosis in Western society has fuelled substantial CFTR research and therapeutic agent development. In general, mutations to CFTR alter cell surface expression, channel gating properties, or both. At present, there is insufficient information to rationally design chemical therapeutics: therefore, high throughput screening, a reliable, sensitive and cost-effective assay to screen libraries of drug-like compounds, is the strategy of choice to identify potential CFTR therapeutics.237 High throughput screens have identified several compounds that reliably enhance CFTR function, although in many cases, the precise mechanisms of action have yet to be elucidated.237
CFTR therapeutics is broadly segregated into two distinct classes: “CFTR potentiators” and “CFTR correctors”. Potentiators (e.g. Ivacaftor/Kalydeco®) enhance channel gating,238 and thus only increase CFTR activity when the channels are present at cell membrane. Consequently, CFTR potentiators display limited efficacy when CFTR abundance is very low.239 In contrast to potentiators, correctors interact with CFTR motifs and/or CFTR interacting proteins that orchestrate and control the processing of CFTR, its delivery to and expression at the apical membrane.240 This therapeutic strategy is termed “proteostasis,”241 because the aim is to treat cystic fibrosis by manipulating the stability and location of CFTR. Lumacaftor (VX-809)222 is the best-characterized corrector compound to date: the FDA recently approved Lumacaftor/Ivacaftor combination therapy (Orkambi™) for treating cystic fibrosis stemming from the ΔF508 mutation (which profoundly disrupts CFTR trafficking and cell surface expression).242 Combining potentiators and correctors (i.e. Orkambi) is an obvious pharmacological strategy that would maximally boost CFTR activity; however, to be viable as an SAH treatment, the respective entities would have to exert effects on non-mutant CFTR, since the wild-type form will be present in the vast majority of patients. Fortunately, both ivacaftor226 and lumacaftor243 exert their respective activity and expression effects on wild-type CFTR.
The availability of CFTR therapeutics that increase wild-type CFTR activity/expression presents an opportunity to repurpose the FDA-approved pharmaceutical Orkambi for SAH therapy. Without doubt, it would be premature to initiate clinical trials prior to undertaking the experimental animal studies. However, in anticipation that CFTR therapeutics will work experimentally, as the strong correlation between CFTR expression and vascular reactivity implies,35 it is tempting to briefly comment on the limited clinical data available with respect to the safety of CFTR therapeutics in patients who do not have cystic fibrosis. At present, clinical trial data for lumacaftor, ivacaftor, and lumacaftor/ivacaftor in healthy subjects are proprietary and therefore, not publicly available. NIH clinical trial records indicate that initial safety studies in healthy subjects were completed in 2010 (NCT00966602) and 2012 (NCT01216046) and included the analyses of laboratory values (clinical chemistry, hematology, coagulation, and urinalysis), electrocardiograms, and vital signs. The fact that four clinical trials involving healthy subjects were subsequently completed (NCT01910415, NCT01768663, NCT01888393 NCT01899105) and a fifth is now underway (NCT02310789) is a strong indication that no serious adverse events have been encountered. It is therefore reasonable to assume that CFTR therapeutics do not pose an overt safety risk to patients with normal CFTR.
Summary
Delayed cerebral ischemia is a primary determinant of clinical outcome in SAH patients fortunate enough to survive the initial hemorrhagic event. Although several factors undoubtedly contribute to this ischemic event, there is little doubt that the principal cause is microvascular dysfunction. This review aims to highlight the fact that, while significant research efforts have been undertaken over the last 20 years, microvascular studies in the SAH field are few, dated and frequently descriptive in nature. In particular, only a couple of investigations have attempted to determine the molecular basis for SAH-induced changes in myogenic reactivity, the primary mechanism of cerebral blood flow autoregulation. This knowledge gap is significant, as it is impossible to develop effective therapeutic interventions without understanding the causal mechanisms. This presumably explains why only one therapeutic intervention, nimodipine, has successfully garnered FDA approval – over 10 years ago. Clearly, further research is needed to fully understand the microvascular mechanisms underlying delayed ischemia in SAH (see Table 1 for a synopsis of proposed microvascular mechanisms and clinical trial outcomes, if applicable). However, given the promising results in recent advancements,35 there is reason to be optimistic that understanding the relevant microvascular mechanisms in SAH will yield effective approaches that specifically correct vascular dysfunction and thereby improve neurological and clinical outcome.
Table 1.
Summary of proposed mechanisms augmenting microvascular constriction in subarachnoid hemorrhage.
Therapeutic target | Proposed pathological mechanism | Supporting references | Clinically utilized drug | Clinical trial outcome | Trial references | Potential issues with intervention |
---|---|---|---|---|---|---|
Endothelium- Dependent Vasodilator Responses | Increased phosphodiesterase type 5 activity attenuates smooth muscle responses to endothelial vasodilators | 146–150 | sildenafil | Improves angiographic vasospasm, but does not increase cerebral perfusion | 155,156 | Non-specifically promotes vasodilation; may disrupt autoregulation. Mechanism not clearly demonstrated to alter myogenic reactivity. |
Potassium Channels | Reduced potassium channel current leads to smooth muscle depolarization/constriction | 89,91,159,160,164,165,169 | CGRP, Retigabine, Celecoxib | CGRP benefit is inconclusive; Retigabine, Celecoxib not yet tested clinically | 170,171 | Non-specifically promotes vasodilation; may disrupt autoregulation. |
20-HETE | Synthesis of pro-constrictive 20-HETE is enhanced | 179,187–189 | – | No interventional clinical trials for this proposed mechanism | – | 20-HETE has many signaling functions beyond vascular tone regulation. |
Tumor necrosis factor | Increased tumor necrosis factor signaling activates pro-constrictive signaling pathways | 35,199,201,208 | etanercept | Trial not started | NCT01865630 | anti-TNF therapy may immunocompromise patients |
S1P receptor 2 subtype | Synthesis of pro-constrictive S1P is enhanced | 35,210,211 | – | No interventional clinical trials for this proposed mechanism | – | No specific S1P2R blockers are clinically available. S1P has many signaling functions beyond vascular tone regulation. |
CFTR | CFTR activity is reduced, which increases pro-constrictive S1P levels | 35,190 | ivacaftor, lumacaftor | No interventional clinical trials for this proposed mechanism | – | Proposed mechanism has not been experimentally demonstrated |
20-HETE: 20 hydroxyeicosatetraenoic acid; CGRP: calcitonin-gene-related peptide; S1P: sphingosine-1-phosphate; S1P2R: S1P receptor 2 subtype; CFTR: cystic fibrosis transmembrane conductance regulator.
Acknowledgement
The authors thank Alexandra Erin Papaelias for graphic design.
Funding
The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: We gratefully acknowledge funding support from a Heart and Stroke Foundation of Canada Grant-in-Aid (StSB; G16-00014175), a Heart and Stroke Foundation of Canada Career Investigator Award (StSB CI-7432) and the Brain Aneurysm Foundation Northwell Health – North Shore University Hospital Brain Aneurysm Centre Chair of Research Award (StSB).
Declaration of conflicting interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Authors’ contributions
All three authors (DL, JTK and StSB) significantly contributed to the conceptualization of this review and the writing/revision process.
References
- 1.Tso MK, Macdonald RL. Subarachnoid hemorrhage: a review of experimental studies on the microcirculation and the neurovascular unit. Transl Stroke Res 2014; 5: 174–189. [DOI] [PubMed] [Google Scholar]
- 2.Solenski NJ, Haley EC, Kassell NF, et al. Medical complications of aneurysmal subarachnoid hemorrhage: a report of the multicenter, cooperative aneurysm study. Participants of the Multicenter Cooperative Aneurysm Study. Crit Care Med 1995; 23: 1007–1017. [DOI] [PubMed] [Google Scholar]
- 3.Hop JW, Rinkel GJ, Algra A, et al. Case-fatality rates and functional outcome after subarachnoid hemorrhage: a systematic review. Stroke 1997; 28: 660–664. [DOI] [PubMed] [Google Scholar]
- 4.Johnston SC, Selvin S, Gress DR. The burden, trends, and demographics of mortality from subarachnoid hemorrhage. Neurology 1998; 50: 1413–1418. [DOI] [PubMed] [Google Scholar]
- 5.van Gijn J, Kerr RS, Rinkel GJ. Subarachnoid haemorrhage. Lancet 2007; 369: 306–318. [DOI] [PubMed] [Google Scholar]
- 6.Brilstra EH, Rinkel GJ, Algra A, et al. Rebleeding, secondary ischemia, and timing of operation in patients with subarachnoid hemorrhage. Neurology 2000; 55: 1656–1660. [DOI] [PubMed] [Google Scholar]
- 7.Macdonald RL. Delayed neurological deterioration after subarachnoid haemorrhage. Nat Rev Neurol 2014; 10: 44–58. [DOI] [PubMed] [Google Scholar]
- 8.Vergouwen MD, Vermeulen M, van Gijn J, 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 2010; 41: 2391–2395. [DOI] [PubMed] [Google Scholar]
- 9.Yanagisawa M, Kurihara H, Kimura S, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 1988; 332: 411–415. [DOI] [PubMed] [Google Scholar]
- 10.Salom JB, Torregrosa G, Alborch E. Endothelins and the cerebral circulation. Cerebrovasc Brain Metab Rev 1995; 7: 131–152. [PubMed] [Google Scholar]
- 11.Seifert V, Löffler BM, Zimmermann M, et al. Endothelin concentrations in patients with aneurysmal subarachnoid hemorrhage. Correlation with cerebral vasospasm, delayed ischemic neurological deficits, and volume of hematoma. J Neurosurg 1995; 82: 55–62. [DOI] [PubMed] [Google Scholar]
- 12.Mascia L, Fedorko L, Stewart DJ, et al. Temporal relationship between endothelin-1 concentrations and cerebral vasospasm in patients with aneurysmal subarachnoid hemorrhage. Stroke 2001; 32: 1185–1190. [DOI] [PubMed] [Google Scholar]
- 13.Juvela S. Plasma endothelin concentrations after aneurysmal subarachnoid hemorrhage. J Neurosurg 2000; 92: 390–400. [DOI] [PubMed] [Google Scholar]
- 14.Laban KG, Vergouwen MD, Dijkhuizen RM, et al. Effect of endothelin receptor antagonists on clinically relevant outcomes after experimental subarachnoid hemorrhage: a systematic review and meta-analysis. J Cereb Blood Flow Metab 2015; 35: 1085–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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]
- 16.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]
- 17.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]
- 18.Etminan N, Vergouwen MD, Ilodigwe D, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rabinstein AA, Friedman JA, Weigand SD, et al. Predictors of cerebral infarction in aneurysmal subarachnoid hemorrhage. Stroke 2004; 35: 1862–1866. [DOI] [PubMed] [Google Scholar]
- 20.Fergusen S, Macdonald RL. Predictors of cerebral infarction in patients with aneurysmal subarachnoid hemorrhage. Neurosurgery 2007; 60: 658–667. [DOI] [PubMed] [Google Scholar]
- 21.Jabbarli R, Reinhard M, Niesen WD, et al. Predictors and impact of early cerebral infarction after aneurysmal subarachnoid hemorrhage. Eur J Neurol 2015; 22: 941–947. [DOI] [PubMed] [Google Scholar]
- 22.Stroke Therapy Academic Industry Roundtable (STAIR). Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke 1999; 30: 2752–2758. [DOI] [PubMed] [Google Scholar]
- 23.Fisher M, Feuerstein G, Howells DW, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke 2009; 40: 2244–2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sena E, van der Worp HB, Howells D, et al. How can we improve the pre-clinical development of drugs for stroke? Trends Neurosci 2007; 30: 433–439. [DOI] [PubMed] [Google Scholar]
- 25.O'Collins VE, Macleod MR, Donnan GA, et al. 1,026 experimental treatments in acute stroke. Ann Neurol 2006; 59: 467–477. [DOI] [PubMed] [Google Scholar]
- 26.Vergouwen MD, Ilodigwe D, Macdonald RL. Cerebral infarction after subarachnoid hemorrhage contributes to poor outcome by vasospasm-dependent and -independent effects. Stroke 2011; 42: 924–929. [DOI] [PubMed] [Google Scholar]
- 27.Adamczyk P, He S, Amar AP, et al. Medical management of cerebral vasospasm following aneurysmal subarachnoid hemorrhage: a review of current and emerging therapeutic interventions. Neurol Res Int 2013; 2013: 462491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Etminan N, Vergouwen MD, Macdonald RL. Angiographic vasospasm versus cerebral infarction as outcome measures after aneurysmal subarachnoid hemorrhage. Acta Neurochir Suppl 2013; 115: 33–40. [DOI] [PubMed] [Google Scholar]
- 29.Dhar R, Scalfani MT, Blackburn S, et al. Relationship between angiographic vasospasm and regional hypoperfusion in aneurysmal subarachnoid hemorrhage. Stroke 2012; 43: 1788–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Heilbrun MP, Olesen J, Lassen NA. Regional cerebral blood flow studies in subarachnoid hemorrhage. J Neurosurg 1972; 37: 36–44. [DOI] [PubMed] [Google Scholar]
- 31.Kelly PJ, Gorten RJ, Grossman RG, et al. Cerebral perfusion, vascular spasm, and outcome in patients with ruptured intracranial aneurysms. J Neurosurg 1977; 47: 44–49. [DOI] [PubMed] [Google Scholar]
- 32.Schubert GA, Seiz M, Hegewald AA, et al. Acute hypoperfusion immediately after subarachnoid hemorrhage: a xenon contrast-enhanced CT study. J Neurotrauma 2009; 26: 2225–2231. [DOI] [PubMed] [Google Scholar]
- 33.Géraud G, Tremoulet M, Guell A, et al. The prognostic value of noninvasive CBF measurement in subarachnoid hemorrhage. Stroke 1984; 15: 301–305. [DOI] [PubMed] [Google Scholar]
- 34.Ohkuma H, Suzuki S, Kudo K, et al. Cortical blood flow during cerebral vasospasm after aneurysmal subarachnoid hemorrhage: three-dimensional N-isopropyl-p-[(123)I]iodoamphetamine single photon emission CT findings. AJNR Am J Neuroradiol 2003; 24: 444–450. [PMC free article] [PubMed] [Google Scholar]
- 35.Yagi K, Lidington D, Wan H, et al. Therapeutically targeting tumor necrosis factor-α/sphingosine-1-phosphate signaling corrects myogenic reactivity in subarachnoid hemorrhage. Stroke 2015; 46: 2260–2270. [DOI] [PubMed] [Google Scholar]
- 36.Ogoh S. Relationship between cognitive function and regulation of cerebral blood flow. J Physiol Sci 2017; 67: 345–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.van Mook WN, Rennenberg RJ, Schurink GW, et al. Cerebral hyperperfusion syndrome. Lancet Neurol 2005; 4: 877–888. [DOI] [PubMed] [Google Scholar]
- 38.Bidani AK, Griffin KA, Williamson G, et al. Protective importance of the myogenic response in the renal circulation. Hypertension 2009; 54: 393–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kono M, Belyantseva IA, Skoura A, et al. Deafness and stria vascularis defects in S1P2 receptor-null mice. J Biol Chem 2007; 282: 10690–10696. [DOI] [PubMed] [Google Scholar]
- 40.Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation. Cerebrovasc Brain Metab Rev 1990; 2: 161–192. [PubMed] [Google Scholar]
- 41.Drummond JC. The lower limit of autoregulation: time to revise our thinking? Anesthesiology 1997; 86: 1431–1433. [DOI] [PubMed] [Google Scholar]
- 42.Lecrux C, Hamel E. The neurovascular unit in brain function and disease. Acta Physiol (Oxf) 2011; 203: 47–59. [DOI] [PubMed] [Google Scholar]
- 43.Phillips AA, Chan FH, Zheng MM, 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]
- 44.Jaeger M, Schuhmann MU, Soehle M, et al. Continuous monitoring of cerebrovascular autoregulation after subarachnoid hemorrhage by brain tissue oxygen pressure reactivity and its relation to delayed cerebral infarction. Stroke 2007; 38: 981–986. [DOI] [PubMed] [Google Scholar]
- 45.Jaeger M, Soehle M, Schuhmann MU, et al. Clinical significance of impaired cerebrovascular autoregulation after severe aneurysmal subarachnoid hemorrhage. Stroke 2012; 43: 2097–2101. [DOI] [PubMed] [Google Scholar]
- 46.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]
- 47.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]
- 48.Davis MJ, Hill MA. Signaling mechanisms underlying the vascular myogenic response. Physiol Rev 1999; 79: 387. [DOI] [PubMed] [Google Scholar]
- 49.Schubert R, Mulvany MJ. The myogenic response: established facts and attractive hypotheses. Clin Sci (Lond) 1999; 96: 313–326. [PubMed] [Google Scholar]
- 50.Hill MA, Zou H, Potocnik SJ, et al. Invited review: arteriolar smooth muscle mechanotransduction: Ca(2+) signaling pathways underlying myogenic reactivity. J Appl Physiol (1985) 2001; 91: 973–983. [DOI] [PubMed] [Google Scholar]
- 51.Hill MA, Davis MJ, Meininger GA, et al. Arteriolar myogenic signalling mechanisms: implications for local vascular function. Clin Hemorheol Microcirc 2006; 34: 67–79. [PubMed] [Google Scholar]
- 52.Schubert R, Lidington D, Bolz SS. The emerging role of Ca2 + sensitivity regulation in promoting myogenic vasoconstriction. Cardiovasc Res 2008; 77: 8–18. [DOI] [PubMed] [Google Scholar]
- 53.Hill MA, Meininger GA, Davis MJ, et al. Therapeutic potential of pharmacologically targeting arteriolar myogenic tone. Trends Pharmacol Sci 2009; 30: 363–374. [DOI] [PubMed] [Google Scholar]
- 54.Baek EB, Kim SJ. Mechanisms of myogenic response: Ca(2+)-dependent and -independent signaling. J Smooth Muscle Res 2011; 47: 55–65. [DOI] [PubMed] [Google Scholar]
- 55.Cole WC, Welsh DG. Role of myosin light chain kinase and myosin light chain phosphatase in the resistance arterial myogenic response to intravascular pressure. Arch Biochem Biophys 2011; 510: 160–173. [DOI] [PubMed] [Google Scholar]
- 56.Lidington D, Schubert R, Bolz SS. Capitalizing on diversity: an integrative approach towards the multiplicity of cellular mechanisms underlying myogenic responsiveness. Cardiovasc Res 2013; 97: 404–412. [DOI] [PubMed] [Google Scholar]
- 57.Kuo L, Chilian WM, Davis MJ. Coronary arteriolar myogenic response is independent of endothelium. Circ Res 1990; 66: 860–866. [DOI] [PubMed] [Google Scholar]
- 58.Falcone JC, Davis MJ, Meininger GA. Endothelial independence of myogenic response in isolated skeletal muscle arterioles. Am J Physiol 1991; 260: H130–H135. [DOI] [PubMed] [Google Scholar]
- 59.Meininger GA, Davis MJ. Cellular mechanisms involved in the vascular myogenic response. Am J Physiol 1992; 263: H647–H659. [DOI] [PubMed] [Google Scholar]
- 60.Zou H, Ratz PH, Hill MA. Temporal aspects of Ca(2+) and myosin phosphorylation during myogenic and norepinephrine-induced arteriolar constriction. J Vasc Res 2000; 37: 556–567. [DOI] [PubMed] [Google Scholar]
- 61.El Beheiry MH, Heximer SP, Voigtlaender-Bolz J, et al. Metoprolol impairs resistance artery function in mice. J Appl Physiol (1985) 2011; 111: 1125–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zou H, Ratz PH, Hill MA. Role of myosin phosphorylation and [Ca2+]i in myogenic reactivity and arteriolar tone. Am J Physiol 1995; 269: H1590–H1596. [DOI] [PubMed] [Google Scholar]
- 63.Davis MJ. Perspective: physiological role(s) of the vascular myogenic response. Microcirculation 2012; 19: 99–114. [DOI] [PubMed] [Google Scholar]
- 64.de Wit C, Jahrbeck B, Schafer C, et al. Nitric oxide opposes myogenic pressure responses predominantly in large arterioles in vivo. Hypertension 1998; 31: 787–794. [DOI] [PubMed] [Google Scholar]
- 65.Mulvany MJ, Aalkjaer C. Structure and function of small arteries. Physiol Rev 1990; 70: 921–961. [DOI] [PubMed] [Google Scholar]
- 66.Brekke JF, Gokina NI, Osol G. Vascular smooth muscle cell stress as a determinant of cerebral artery myogenic tone. Am J Physiol Heart Circ Physiol 2002; 283: H2210–H2216. [DOI] [PubMed] [Google Scholar]
- 67.Davis MJ. Myogenic response gradient in an arteriolar network. Am J Physiol 1993; 264: H2168–H2179. [DOI] [PubMed] [Google Scholar]
- 68.Metting PJ, Stein PM, Stoos BA, et al. Systemic vascular autoregulation amplifies pressor responses to vasoconstrictor agents. Am J Physiol 1989; 256: R98–105. [DOI] [PubMed] [Google Scholar]
- 69.Metting PJ, Kostrzewski KA, Stein PM, et al. Quantitative contribution of systemic vascular autoregulation in acute hypertension in conscious dogs. J Clin Invest 1989; 84: 1900–1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Moien-Afshari F, Skarsgard PL, McManus BM, et al. Cardiac transplantation and resistance artery myogenic tone. Can J Physiol Pharmacol 2004; 82: 840–848. [DOI] [PubMed] [Google Scholar]
- 71.Spassova MA, Hewavitharana T, Xu W, et al. A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc Natl Acad Sci U S A 2006; 103: 16586–16591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Welsh DG, Morielli AD, Nelson MT, et al. Transient receptor potential channels regulate myogenic tone of resistance arteries. Circ Res 2002; 90: 248–250. [DOI] [PubMed] [Google Scholar]
- 73.Wang Y, Deng X, Hewavitharana T, et al. Stim, ORAI and TRPC channels in the control of calcium entry signals in smooth muscle. Clin Exp Pharmacol Physiol 2008; 35: 1127–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mederos y Schnitzler M, Storch U, Meibers S, et al. Gq-coupled receptors as mechanosensors mediating myogenic vasoconstriction. EMBO J 2008; 27: 3092–3103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Martinez-Lemus LA, Crow T, Davis MJ, et al. alphavbeta3- and alpha5beta1-integrin blockade inhibits myogenic constriction of skeletal muscle resistance arterioles. Am J Physiol Heart Circ Physiol 2005; 289: H322–H329. [DOI] [PubMed] [Google Scholar]
- 76.Sun Z, Martinez-Lemus LA, Hill MA, et al. Extracellular matrix-specific focal adhesions in vascular smooth muscle produce mechanically active adhesion sites. Am J Physiol Cell Physiol 2008; 295: C268–C278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kroetsch JT, Levy AS, Zhang H, et al. Constitutive smooth muscle tumour necrosis factor regulates microvascular myogenic responsiveness and systemic blood pressure. Nat Commun 2017; 8: 14805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Somlyo AP, Somlyo AV. Ca2 + sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003; 83: 1325–1358. [DOI] [PubMed] [Google Scholar]
- 79.Kotecha N, Hill MA. Myogenic contraction in rat skeletal muscle arterioles: smooth muscle membrane potential and Ca(2+) signaling. Am J Physiol Heart Circ Physiol 2005; 289: H1326–H1334. [DOI] [PubMed] [Google Scholar]
- 80.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]
- 81.Knot HJ, Standen NB, Nelson MT. Ryanodine receptors regulate arterial diameter and wall [Ca2+] in cerebral arteries of rat via Ca2+-dependent K + channels. J Physiol 1998; 508: 211–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Watanabe J, Karibe A, Horiguchi S, et al. Modification of myogenic intrinsic tone and [Ca2+]i of rat isolated arterioles by ryanodine and cyclopiazonic acid. Circ Res 1993; 73: 465–472. [DOI] [PubMed] [Google Scholar]
- 83.Johnson RP, El-Yazbi AF, Takeya K, et al. Ca2 + sensitization via phosphorylation of myosin phosphatase targeting subunit at threonine-855 by Rho kinase contributes to the arterial myogenic response. J Physiol 2009; 587: 2537–2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.El-Yazbi AF, Johnson RP, Walsh EJ, et al. Pressure-dependent contribution of Rho kinase-mediated calcium sensitization in serotonin-evoked vasoconstriction of rat cerebral arteries. J Physiol 2010; 588: 1747–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bolz SS, Vogel L, Sollinger D, et al. Sphingosine kinase modulates microvascular tone and myogenic responses through activation of RhoA/Rho kinase. Circulation 2003; 108: 342–347. [DOI] [PubMed] [Google Scholar]
- 86.Korzick DH, Laughlin MH, Bowles DK. Alterations in PKC signaling underlie enhanced myogenic tone in exercise-trained porcine coronary resistance arteries. J Appl Physiol (1985) 2004; 96: 1425–1432. [DOI] [PubMed] [Google Scholar]
- 87.Dessy C, Matsuda N, Hulvershorn J, et al. Evidence for involvement of the PKC-alpha isoform in myogenic contractions of the coronary microcirculation. Am J Physiol Heart Circ Physiol 2000; 279: H916–H923. [DOI] [PubMed] [Google Scholar]
- 88.Ursino M. Mechanisms of cerebral blood flow regulation. Crit Rev Biomed Eng 1991; 18: 255–288. [PubMed] [Google Scholar]
- 89.Nystoriak MA, O'Connor KP, Sonkusare SK, et al. Fundamental increase in pressure-dependent constriction of brain parenchymal arterioles from subarachnoid hemorrhage model rats due to membrane depolarization. Am J Physiol Heart Circ Physiol 2011; 300: H803–H812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wellman GC, Koide M. Impact of subarachnoid hemorrhage on parenchymal arteriolar function. Acta Neurochir Suppl 2013; 115: 173–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ishiguro M, Puryear CB, Bisson E, et al. Enhanced myogenic tone in cerebral arteries from a rabbit model of subarachnoid hemorrhage. Am J Physiol Heart Circ Physiol 2002; 283: H2217–H2225. [DOI] [PubMed] [Google Scholar]
- 92.Koide M, Nystoriak MA, Brayden JE, et al. Impact of subarachnoid hemorrhage on local and global calcium signaling in cerebral artery myocytes. Acta Neurochir Suppl 2011; 110: 145–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kim P, Sundt TM, Vanhoutte PM. Alterations of mechanical properties in canine basilar arteries after subarachnoid hemorrhage. J Neurosurg 1989; 71: 430–436. [DOI] [PubMed] [Google Scholar]
- 94.Yamaguchi-Okada M, Nishizawa S, Koide M, et al. Biomechanical and phenotypic changes in the vasospastic canine basilar artery after subarachnoid hemorrhage. J Appl Physiol (1985) 2005; 99: 2045–2052. [DOI] [PubMed] [Google Scholar]
- 95.Lin CL, Calisaneller T, Ukita N, et al. A murine model of subarachnoid hemorrhage-induced cerebral vasospasm. J Neurosci Meth 2003; 123: 89–97. [DOI] [PubMed] [Google Scholar]
- 96.Barone FC, Knudsen DJ, Nelson AH, et al. Mouse strain differences in susceptibility to cerebral ischemia are related to cerebral vascular anatomy. J Cereb Blood Flow Metab 1993; 13: 683–692. [DOI] [PubMed] [Google Scholar]
- 97.Fujii M, Hara H, Meng W, et al. Strain-related differences in susceptibility to transient forebrain ischemia in SV-129 and C57black/6 mice. Stroke 1997; 28: 1805–1810. [DOI] [PubMed] [Google Scholar]
- 98.Baron JC. Perfusion thresholds in human cerebral ischemia: historical perspective and therapeutic implications. Cerebrovasc Dis 2001; 11: 2–8. [DOI] [PubMed] [Google Scholar]
- 99.Schlaug G, Benfield A, Baird AE, et al. The ischemic penumbra: operationally defined by diffusion and perfusion MRI. Neurology 1999; 53: 1528–1537. [DOI] [PubMed] [Google Scholar]
- 100.Røhl L, Ostergaard L, Simonsen CZ, et al. Viability thresholds of ischemic penumbra of hyperacute stroke defined by perfusion-weighted MRI and apparent diffusion coefficient. Stroke 2001; 32: 1140–1146. [DOI] [PubMed] [Google Scholar]
- 101.Liu Y, Karonen JO, Vanninen RL, et al. Cerebral hemodynamics in human acute ischemic stroke: a study with diffusion- and perfusion-weighted magnetic resonance imaging and SPECT. J Cereb Blood Flow Metab 2000; 20: 910–920. [DOI] [PubMed] [Google Scholar]
- 102.Velat GJ, Kimball MM, Mocco JD, et al. Vasospasm after aneurysmal subarachnoid hemorrhage: review of randomized controlled trials and meta-analyses in the literature. World Neurosurg 2011; 76: 446–454. [DOI] [PubMed] [Google Scholar]
- 103.Sen J, Belli A, Albon H, et al. Triple-H therapy in the management of aneurysmal subarachnoid haemorrhage. Lancet Neurol 2003; 2: 614–621. [DOI] [PubMed] [Google Scholar]
- 104.Lee KH, Lukovits T, Friedman JA. “Triple-H” therapy for cerebral vasospasm following subarachnoid hemorrhage. Neurocrit Care 2006; 4: 68–76. [DOI] [PubMed] [Google Scholar]
- 105.Nornes H, Knutzen HB, Wikeby P. Cerebral arterial blood flow and aneurysm surgery. Part 2: Induced hypotension and autoregulatory capacity. J Neurosurg 1977; 47: 819–827. [DOI] [PubMed] [Google Scholar]
- 106.Yamamoto S, Nishizawa S, Tsukada H, et al. Cerebral blood flow autoregulation following subarachnoid hemorrhage in rats: chronic vasospasm shifts the upper and lower limits of the autoregulatory range toward higher blood pressures. Brain Res 1998; 782: 194–201. [DOI] [PubMed] [Google Scholar]
- 107.Lennihan L, Mayer SA, Fink ME, et al. Effect of hypervolemic therapy on cerebral blood flow after subarachnoid hemorrhage: a randomized controlled trial. Stroke 2000; 31: 383–391. [DOI] [PubMed] [Google Scholar]
- 108.Egge A, Waterloo K, Sjøholm H, et al. Prophylactic hyperdynamic postoperative fluid therapy after aneurysmal subarachnoid hemorrhage: a clinical, prospective, randomized, controlled study. Neurosurgery 2001; 49: 593–605. [DOI] [PubMed] [Google Scholar]
- 109.Dankbaar JW, Slooter AJ, Rinkel GJ, et al. Effect of different components of triple-H therapy on cerebral perfusion in patients with aneurysmal subarachnoid haemorrhage: a systematic review. Crit Care 2010; 14: R23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Connolly ES, Rabinstein AA, Carhuapoma JR, et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage: a guideline for healthcare professionals from the American Heart Association/american Stroke Association. Stroke 2012; 43: 1711–1737. [DOI] [PubMed] [Google Scholar]
- 111.Ray WZ, Moran CJ, Derdeyn CP, et al. Near-complete resolution of angiographic cerebral vasospasm after extreme elevation of mean arterial pressure: case report. Surg Neurol 2009; 72: 347–353. [DOI] [PubMed] [Google Scholar]
- 112.Ogungbo B, Prakash S, Ushewokunze S, et al. Value of triple H therapy in a patient with an ischemic penumbra following subarachnoid hemorrhage: a case study. J Neurosci Nurs 2005; 37: 326–328. [DOI] [PubMed] [Google Scholar]
- 113.Meyer R, Deem S, Yanez ND, et al. Current practices of triple-H prophylaxis and therapy in patients with subarachnoid hemorrhage. Neurocrit Care 2011; 14: 24–36. [DOI] [PubMed] [Google Scholar]
- 114.Mortimer AM, Steinfort B, Faulder K, et al. The detrimental clinical impact of severe angiographic vasospasm may be diminished by maximal medical therapy and intensive endovascular treatment. J Neurointerv Surg 2015; 7: 881–887. [DOI] [PubMed] [Google Scholar]
- 115.Rosenwasser RH, Armonda RA, Thomas JE, et al. Therapeutic modalities for the management of cerebral vasospasm: timing of endovascular options. Neurosurgery 1999; 44: 975–979. [DOI] [PubMed] [Google Scholar]
- 116.Zwienenberg-Lee M, Hartman J, Rudisill N, et al. Effect of prophylactic transluminal balloon angioplasty on cerebral vasospasm and outcome in patients with Fisher grade III subarachnoid hemorrhage: results of a phase II multicenter, randomized, clinical trial. Stroke 2008; 39: 1759–1765. [DOI] [PubMed] [Google Scholar]
- 117.Petruk KC, West M, Mohr G, et al. Nimodipine treatment in poor-grade aneurysm patients. Results of a multicenter double-blind placebo-controlled trial. J Neurosurg 1988; 68: 505–517. [DOI] [PubMed] [Google Scholar]
- 118.Pickard JD, Murray GD, Illingworth R, et al. Effect of oral nimodipine on cerebral infarction and outcome after subarachnoid haemorrhage: British aneurysm nimodipine trial. BMJ 1989; 298: 636–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Liu GJ, Luo J, Zhang LP, et al. Meta-analysis of the effectiveness and safety of prophylactic use of nimodipine in patients with an aneurysmal subarachnoid haemorrhage. CNS Neurol Disord Drug Targets 2011; 10: 834–844. [DOI] [PubMed] [Google Scholar]
- 120.Espinosa F, Weir B, Overton T, et al. A randomized placebo-controlled double-blind trial of nimodipine after SAH in monkeys. Part 1: Clinical and radiological findings. J Neurosurg 1984; 60: 1167–1175. [DOI] [PubMed] [Google Scholar]
- 121.Philippon J, Grob R, Dagreou F, et al. Prevention of vasospasm in subarachnoid haemorrhage. A controlled study with nimodipine. Acta Neurochir (Wien) 1986; 82: 110–114. [DOI] [PubMed] [Google Scholar]
- 122.Mee E, Dorrance D, Lowe D, et al. Controlled study of nimodipine in aneurysm patients treated early after subarachnoid hemorrhage. Neurosurgery 1988; 22: 484–491. [DOI] [PubMed] [Google Scholar]
- 123.Nuglisch J, Karkoutly C, Mennel HD, et al. Protective effect of nimodipine against ischemic neuronal damage in rat hippocampus without changing postischemic cerebral blood flow. J Cereb Blood Flow Metab 1990; 10: 654–659. [DOI] [PubMed] [Google Scholar]
- 124.Di Mascio R, Marchioli R, Tognoni G. From pharmacological promises to controlled clinical trials to meta-analysis and back: the case of nimodipine in cerebrovascular disorders. Clin Trials Metaanal 1994; 29: 57–79. [PubMed] [Google Scholar]
- 125.Pisani A, Calabresi P, Tozzi A, et al. L-type Ca2 + channel blockers attenuate electrical changes and Ca2 + rise induced by oxygen/glucose deprivation in cortical neurons. Stroke 1998; 29: 196–201. [DOI] [PubMed] [Google Scholar]
- 126.Herzfeld E, Strauss C, Simmermacher S, et al. Investigation of the neuroprotective impact of nimodipine on Neuro2a cells by means of a surgery-like stress model. Int J Mol Sci 2014; 15: 18453–18465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ricci A, Sabbatini M, Tomassoni D, et al. Neuronal populations of rat cerebral cortex and hippocampus expressed a higher density of L-type Ca 2 + channel than corresponding cerebral vessels. Clin Exp Hypertens 2002; 24: 715–726. [DOI] [PubMed] [Google Scholar]
- 128.Hui C, Lau KP. Efficacy of intra-arterial nimodipine in the treatment of cerebral vasospasm complicating subarachnoid haemorrhage. Clin Radiol 2005; 60: 1030–1036. [DOI] [PubMed] [Google Scholar]
- 129.Kerz T, Boor S, Beyer C, et al. Effect of intraarterial papaverine or nimodipine on vessel diameter in patients with cerebral vasospasm after subarachnoid hemorrhage. Br J Neurosurg 2012; 26: 517–524. [DOI] [PubMed] [Google Scholar]
- 130.Kim SS, Park DH, Lim DJ, et al. Angiographic features and clinical outcomes of intra-arterial nimodipine injection in patients with subarachnoid hemorrhage-induced vasospasm. J Korean Neurosurg Soc 2012; 52: 172–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Dorhout Mees SM, Algra A, Vandertop WP, et al. Magnesium for aneurysmal subarachnoid haemorrhage (MASH-2): a randomised placebo-controlled trial. Lancet 2012; 380: 44–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Golan E, Vasquez DN, Ferguson ND, et al. Prophylactic magnesium for improving neurologic outcome after aneurysmal subarachnoid hemorrhage: systematic review and meta-analysis. J Crit Care 2013; 28: 173–181. [DOI] [PubMed] [Google Scholar]
- 133.Haley EC, Kassell NF, Torner JC. A randomized controlled trial of high-dose intravenous nicardipine in aneurysmal subarachnoid hemorrhage. A report of the Cooperative Aneurysm Study. J Neurosurg 1993; 78: 537–547. [DOI] [PubMed] [Google Scholar]
- 134.Huang RQ, Jiang FG, Feng ZM, et al. Nicardipine in the treatment of aneurysmal subarachnoid haemorrhage: a meta-analysis of published data. Acta Neurol Belg 2013; 113: 3–6. [DOI] [PubMed] [Google Scholar]
- 135.Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke 1991; 22: 971–982. [DOI] [PubMed] [Google Scholar]
- 136.Osaka K. Prolonged vasospasm produced by the breakdown products of erythrocytes. J Neurosurg 1977; 47: 403–411. [DOI] [PubMed] [Google Scholar]
- 137.Dora E, Feher E, Farago M, et al. Mechanism of hemoglobin-induced spasm in the isolated middle cerebral artery of the cat. Adv Exp Med Biol 1989; 248: 533–542. [DOI] [PubMed] [Google Scholar]
- 138.Mayberg MR, Okada T, Bark DH. The role of hemoglobin in arterial narrowing after subarachnoid hemorrhage. J Neurosurg 1990; 72: 634–640. [DOI] [PubMed] [Google Scholar]
- 139.Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to subarachnoid hemorrhage visualized by computerized tomographic scanning. Neurosurgery 1980; 6: 1–9. [DOI] [PubMed] [Google Scholar]
- 140.Claassen J, Bernardini GL, Kreiter K, et al. Effect of cisternal and ventricular blood on risk of delayed cerebral ischemia after subarachnoid hemorrhage: the Fisher scale revisited. Stroke 2001; 32: 2012–2020. [DOI] [PubMed] [Google Scholar]
- 141.Sercombe R, Dinh YR, Gomis P. Cerebrovascular inflammation following subarachnoid hemorrhage. Jpn J Pharmacol 2002; 88: 227–249. [DOI] [PubMed] [Google Scholar]
- 142.Siuta M, Zuckerman SL, Mocco J. Nitric oxide in cerebral vasospasm: theories, measurement, and treatment. Neurol Res Int 2013; 2013: 972417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wu HJ, Wu C, Niu HJ, et al. Neuroprotective mechanisms of melatonin in hemorrhagic stroke. Cell Mol Neurobiol 2017. (in press). DOI: 10.1007/s10571-017-0461-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lucke-Wold BP, Logsdon AF, Manoranjan B, et al. Aneurysmal subarachnoid hemorrhage and neuroinflammation: a comprehensive review. Int J Mol Sci 2016; 17: 497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Fuwa I, Mayberg M, Gadjusek C, et al. Enhanced secretion of endothelin by endothelial cells in response to hemoglobin. Neurol Med Chir (Tokyo) 1993; 33: 739–743. [DOI] [PubMed] [Google Scholar]
- 146.Veldeman M, Höllig A, Clusmann H, et al. Delayed cerebral ischaemia prevention and treatment after aneurysmal subarachnoid haemorrhage: a systematic review. Br J Anaesth 2016; 117: 17–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Naraoka M, Matsuda N, Shimamura N, et al. The role of arterioles and the microcirculation in the development of vasospasm after aneurysmal SAH. Biomed Res Int 2014; 2014: 253746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Terpolilli NA, Brem C, Bühler D, et al. Are we barking up the wrong vessels? Cerebral microcirculation after subarachnoid hemorrhage. Stroke 2015; 46: 3014–3019. [DOI] [PubMed] [Google Scholar]
- 149.Pool JL. Cerebral vasospasm. N Engl J Med 1958; 259: 1259–1264. [DOI] [PubMed] [Google Scholar]
- 150.Stornelli SA, French JD. Subarachnoid hemorrhage – Factors in prognosis and management. J Neurosurg 1964; 21: 769–780. [DOI] [PubMed] [Google Scholar]
- 151.Kim P, Lorenz RR, Sundt TM, et al. Release of endothelium-derived relaxing factor after subarachnoid hemorrhage. J Neurosurg 1989; 70: 108–114. [DOI] [PubMed] [Google Scholar]
- 152.Kim P, Schini VB, Sundt TM, et al. Reduced production of cGMP underlies the loss of endothelium-dependent relaxations in the canine basilar artery after subarachnoid hemorrhage. Circ Res 1992; 70: 248–256. [DOI] [PubMed] [Google Scholar]
- 153.Sobey CG, Quan L. Impaired cerebral vasodilator responses to NO and PDE V inhibition after subarachnoid hemorrhage. Am J Physiol 1999; 277: H1718–H1724. [DOI] [PubMed] [Google Scholar]
- 154.Park IS, Meno JR, Witt CE, et al. Impairment of intracerebral arteriole dilation responses after subarachnoid hemorrhage. Laboratory investigation. J Neurosurg 2009; 111: 1008–1013. [DOI] [PubMed] [Google Scholar]
- 155.Onoue H, Kaito N, Akiyama M, et al. Altered reactivity of human cerebral arteries after subarachnoid hemorrhage. J Neurosurg 1995; 83: 510–515. [DOI] [PubMed] [Google Scholar]
- 156.Hatake K, Wakabayashi I, Kakishita E, et al. Impairment of endothelium-dependent relaxation in human basilar artery after subarachnoid hemorrhage. Stroke 1992; 23: 1111–1116. [DOI] [PubMed] [Google Scholar]
- 157.Bergström A, Staalsø JM, Romner B, et al. Impaired endothelial function after aneurysmal subarachnoid haemorrhage correlates with arginine:asymmetric dimethylarginine ratio. Br J Anaesth 2014; 112: 311–318. [DOI] [PubMed] [Google Scholar]
- 158.Kanamaru K, Weir BK, Findlay JM, et al. Pharmacological studies on relaxation of spastic primate cerebral arteries in subarachnoid hemorrhage. J Neurosurg 1989; 71: 909–915. [DOI] [PubMed] [Google Scholar]
- 159.Katusić ZS, Milde JH, Cosentino F, et al. Subarachnoid hemorrhage and endothelial L-arginine pathway in small brain stem arteries in dogs. Stroke 1993; 24: 392–399. [DOI] [PubMed] [Google Scholar]
- 160.Washington CW, Derdeyn CP, Dhar R, et al. A Phase I proof-of-concept and safety trial of sildenafil to treat cerebral vasospasm following subarachnoid hemorrhage. J Neurosurg 2016; 124: 318–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Dhar R, Washington C, Diringer M, et al. Acute effect of intravenous sildenafil on cerebral blood flow in patients with vasospasm after subarachnoid hemorrhage. Neurocrit Care 2016; 25: 201–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Nelson MT, Patlak JB, Worley JF, et al. Calcium channels, potassium channels, and voltage dependence of arterial smooth muscle tone. Am J Physiol 1990; 259: C3–C18. [DOI] [PubMed] [Google Scholar]
- 163.Faraci FM, Sobey CG. Role of potassium channels in regulation of cerebral vascular tone. J Cereb Blood Flow Metab 1998; 18: 1047–1063. [DOI] [PubMed] [Google Scholar]
- 164.Jahromi BS, Aihara Y, Ai J, et al. Voltage-gated K + channel dysfunction in myocytes from a dog model of subarachnoid hemorrhage. J Cereb Blood Flow Metab 2008; 28: 797–811. [DOI] [PubMed] [Google Scholar]
- 165.Harder DR, Dernbach P, Waters A. Possible cellular mechanism for cerebral vasospasm after experimental subarachnoid hemorrhage in the dog. J Clin Invest 1987; 80: 875–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Ishiguro M, Morielli AD, Zvarova K, et al. Oxyhemoglobin-induced suppression of voltage-dependent K + channels in cerebral arteries by enhanced tyrosine kinase activity. Circ Res 2006; 99: 1252–1260. [DOI] [PubMed] [Google Scholar]
- 167.Koide M, Wellman GC. SAH-induced suppression of voltage-gated K(+) (K (V)) channel currents in parenchymal arteriolar myocytes involves activation of the HB-EGF/EGFR pathway. Acta Neurochir Suppl 2013; 115: 179–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Koide M, Wellman GC. SAH-induced MMP activation and K V current suppression is mediated via both ROS-dependent and ROS-independent mechanisms. Acta Neurochir Suppl 2015; 120: 89–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Aihara Y, Jahromi BS, Yassari R, et al. Molecular profile of vascular ion channels after experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2004; 24: 75–83. [DOI] [PubMed] [Google Scholar]
- 170.Mani BK, O'Dowd J, Kumar L, et al. Vascular KCNQ (Kv7) potassium channels as common signaling intermediates and therapeutic targets in cerebral vasospasm. J Cardiovasc Pharmacol 2013; 61: 51–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Jahromi BS, Aihara Y, Ai J, et al. Preserved BK channel function in vasospastic myocytes from a dog model of subarachnoid hemorrhage. J Vasc Res 2008; 45: 402–415. [DOI] [PubMed] [Google Scholar]
- 172.Zhong XZ, Harhun MI, Olesen SP, et al. Participation of KCNQ (Kv7) potassium channels in myogenic control of cerebral arterial diameter. J Physiol 2010; 588: 3277–3293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Kleppisch T, Nelson MT. ATP-sensitive K + currents in cerebral arterial smooth muscle: pharmacological and hormonal modulation. Am J Physiol 1995; 269: H1634–H1640. [DOI] [PubMed] [Google Scholar]
- 174.Flynn LMC, Begg CJ, Macleod MR, et al. Alpha calcitonin gene-related peptide increases cerebral vessel diameter in animal models of subarachnoid hemorrhage: a systematic review and meta-analysis. Front Neurol 2017; 8: 357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Johnston FG, Bell BA, Robertson IJ, et al. Effect of calcitonin-gene-related peptide on postoperative neurological deficits after subarachnoid haemorrhage. Lancet 1990; 335: 869–872. [DOI] [PubMed] [Google Scholar]
- 176.Effect of calcitonin-gene-related peptide in patients with delayed postoperative cerebral ischaemia after aneurysmal subarachnoid haemorrhage. European CGRP in Subarachnoid Haemorrhage Study Group. Lancet 1992; 339: 831–834. [PubMed] [Google Scholar]
- 177.Roman RJ. P-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol Rev 2002; 82: 131–185. [DOI] [PubMed] [Google Scholar]
- 178.Wu CC, Gupta T, Garcia V, et al. 20-HETE and blood pressure regulation: clinical implications. Cardiol Rev 2014; 22: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Pratt PF, Medhora M, Harder DR. Mechanisms regulating cerebral blood flow as therapeutic targets. Curr Opin Investigat Drugs 2004; 5: 952–956. [PubMed] [Google Scholar]
- 180.Imig JD, Simpkins AN, Renic M, et al. Cytochrome P450 eicosanoids and cerebral vascular function. Expert Rev Mol Med 2011; 13: e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.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]
- 182.Gebremedhin D, Gopalakrishnan S, Harder DR. Endogenous events modulating myogenic regulation of cerebrovascular function. Curr Vasc Pharmacol 2014; 12: 810–817. [DOI] [PubMed] [Google Scholar]
- 183.Berg RM. Myogenic and metabolic feedback in cerebral autoregulation: putative involvement of arachidonic acid-dependent pathways. Med Hypotheses 2016; 92: 12–17. [DOI] [PubMed] [Google Scholar]
- 184.Roman RJ, Renic M, Dunn KM, et al. Evidence that 20-HETE contributes to the development of acute and delayed cerebral vasospasm. Neurol Res 2006; 28: 738–749. [DOI] [PubMed] [Google Scholar]
- 185.Gewirtz RJ, Dhillon HS, Goes SE, et al. Lactate and free fatty acids after subarachnoid hemorrhage. Brain Res 1999; 840: 84–91. [DOI] [PubMed] [Google Scholar]
- 186.Pilitsis JG, Coplin WM, O'Regan MH, et al. Measurement of free fatty acids in cerebrospinal fluid from patients with hemorrhagic and ischemic stroke. Brain Res 2003; 985: 198–201. [DOI] [PubMed] [Google Scholar]
- 187.Pilitsis JG, Coplin WM, O'Regan MH, et al. Free fatty acids in human cerebrospinal fluid following subarachnoid hemorrhage and their potential role in vasospasm: a preliminary observation. J Neurosurg 2002; 97: 272–279. [DOI] [PubMed] [Google Scholar]
- 188.Poloyac SM, Reynolds RB, Yonas H, et al. Identification and quantification of the hydroxyeicosatetraenoic acids, 20-HETE and 12-HETE, in the cerebrospinal fluid after subarachnoid hemorrhage. J Neurosci Meth 2005; 144: 257–263. [DOI] [PubMed] [Google Scholar]
- 189.Crago EA, Thampatty BP, Sherwood PR, et al. Cerebrospinal fluid 20-HETE is associated with delayed cerebral ischemia and poor outcomes after aneurysmal subarachnoid hemorrhage. Stroke 2011; 42: 1872–1877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Donnelly MK, Crago EA, Conley YP, et al. 20-HETE is associated with unfavorable outcomes in subarachnoid hemorrhage patients. J Cereb Blood Flow Metab 2015; 35: 1515–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Siler DA, Martini RP, Ward JP, et al. Protective role of p450 epoxyeicosanoids in subarachnoid hemorrhage. Neurocrit Care 2015; 22: 306–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Takeuchi K, Renic M, Bohman QC, et al. Reversal of delayed vasospasm by an inhibitor of the synthesis of 20-HETE. Am J Physiol Heart Circ Physiol 2005; 289: H2203–H2211. [DOI] [PubMed] [Google Scholar]
- 193.Hacein-Bey L, Harder DR, Meier HT, et al. Reversal of delayed vasospasm by TS-011 in the dual hemorrhage dog model of subarachnoid hemorrhage. AJNR Am J Neuroradiol 2006; 27: 1350–1354. [PMC free article] [PubMed] [Google Scholar]
- 194.Kehl F, Cambj-Sapunar L, Maier KG, et al. 20-HETE contributes to the acute fall in cerebral blood flow after subarachnoid hemorrhage in the rat. Am J Physiol Heart Circ Physiol 2002; 282: H1556–H1565. [DOI] [PubMed] [Google Scholar]
- 195.Meissner A, Yang J, Kroetsch JT, et al. Tumor necrosis factor-α-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation 2012; 125: 2739–2750. [DOI] [PubMed] [Google Scholar]
- 196.Yang J, Hossein Noyan-Ashraf M, Meissner A, et al. Proximal cerebral arteries develop myogenic responsiveness in heart failure via tumor necrosis factor-α-dependent activation of sphingosine-1-phosphate signaling. Circulation 2012; 126: 196–206. [DOI] [PubMed] [Google Scholar]
- 197.Lidington D, Peter BF, Meissner A, et al. The phosphorylation motif at serine 225 governs the localization and function of sphingosine kinase 1 in resistance arteries. Arterioscler Thromb Vasc Biol 2009; 29: 1916–1922. [DOI] [PubMed] [Google Scholar]
- 198.Peter BF, Lidington D, Harada A, et al. Role of sphingosine-1-phosphate phosphohydrolase 1 in the regulation of resistance artery tone. Circ Res 2008; 103: 315–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Keller M, Lidington D, Vogel L, et al. Sphingosine kinase functionally links elevated transmural pressure and increased reactive oxygen species formation in resistance arteries. FASEB J 2006; 20: 702–704. [DOI] [PubMed] [Google Scholar]
- 200.Kroetsch JT and Bolz SS The TNF-α/sphingosine-1-phosphate signaling axis drives myogenic responsiveness in heart failure. J Vasc Res 2013; 50: 177–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Mathiesen T, Edner G, Ulfarsson E, et al. Cerebrospinal fluid interleukin-1 receptor antagonist and tumor necrosis factor-alpha following subarachnoid hemorrhage. J Neurosurg 1997; 87: 215–220. [DOI] [PubMed] [Google Scholar]
- 202.Hanafy KA, Grobelny B, Fernandez L, et al. Brain interstitial fluid TNF-alpha after subarachnoid hemorrhage. J Neurol Sci 2010; 291: 69–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Lad SP, Hegen H, Gupta G, et al. Proteomic biomarker discovery in cerebrospinal fluid for cerebral vasospasm following subarachnoid hemorrhage. J Stroke Cerebrovasc Dis 2012; 21: 30–41. [DOI] [PubMed] [Google Scholar]
- 204.Fassbender K, Hodapp B, Rossol S, et al. Inflammatory cytokines in subarachnoid haemorrhage: association with abnormal blood flow velocities in basal cerebral arteries. J Neurol Neurosurg Psychiatry 2001; 70: 534–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Hanafy KA, Stuart RM, Khandji AG, et al. Relationship between brain interstitial fluid tumor necrosis factor-α and cerebral vasospasm after aneurysmal subarachnoid hemorrhage. J Clin Neurosci 2010; 17: 853–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Vecchione C, Frati A, Di Pardo A, et al. Tumor necrosis factor-alpha mediates hemolysis-induced vasoconstriction and the cerebral vasospasm evoked by subarachnoid hemorrhage. Hypertension 2009; 54: 150–156. [DOI] [PubMed] [Google Scholar]
- 207.Lim M, Choi SK, Cho YE, et al. The role of sphingosine kinase 1/sphingosine-1-phosphate pathway in the myogenic tone of posterior cerebral arteries. PLoS One 2012; 7: e35177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Salomone S, Soydan G, Ip PC, et al. Vessel-specific role of sphingosine kinase 1 in the vasoconstriction of isolated basilar arteries. Pharmacol Res 2010; 62: 465–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Tosaka M, Okajima F, Hashiba Y, et al. Sphingosine 1-phosphate contracts canine basilar arteries in vitro and in vivo: possible role in pathogenesis of cerebral vasospasm. Stroke 2001; 32: 2913–2919. [DOI] [PubMed] [Google Scholar]
- 210.Coussin F, Scott RH, Wise A, et al. Comparison of sphingosine 1-phosphate-induced intracellular signaling pathways in vascular smooth muscles: differential role in vasoconstriction. Circ Res 2002; 91: 151–157. [DOI] [PubMed] [Google Scholar]
- 211.Murakami A, Takasugi H, Ohnuma S, et al. Sphingosine 1-phosphate (S1P) regulates vascular contraction via S1P3 receptor: investigation based on a new S1P3 receptor antagonist. Mol Pharmacol 2010; 77: 704–713. [DOI] [PubMed] [Google Scholar]
- 212.Coussin F, Scott RH, Nixon GF. Sphingosine 1-phosphate induces CREB activation in rat cerebral artery via a protein kinase C-mediated inhibition of voltage-gated K + channels. Biochem Pharmacol 2003; 66: 1861–1870. [DOI] [PubMed] [Google Scholar]
- 213.Zhang BF, Song JN, Ma XD, et al. Etanercept alleviates early brain injury following experimental subarachnoid hemorrhage and the possible role of tumor necrosis factor-α and c-Jun N-terminal kinase pathway. Neurochem Res 2015; 40: 591–599. [DOI] [PubMed] [Google Scholar]
- 214.Bigaud M, Guerini D, Billich A, et al. Second generation S1P pathway modulators: research strategies and clinical developments. Biochim Biophys Acta 2014; 1841: 745–758. [DOI] [PubMed] [Google Scholar]
- 215.Xu HL, Pelligrino DA, Paisansathan C, et al. Protective role of fingolimod (FTY720) in rats subjected to subarachnoid hemorrhage. J Neuroinflammation 2015; 12: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Fu Y, Hao J, Zhang N, et al. Fingolimod for the treatment of intracerebral hemorrhage: a 2-arm proof-of-concept study. JAMA Neurol 2014; 71: 1092–1101. [DOI] [PubMed] [Google Scholar]
- 217.Murdaca G, Spanò F, Contatore M, et al. Infection risk associated with anti-TNF-α agents: a review. Expert Opin Drug Saf 2015; 14: 571–582. [DOI] [PubMed] [Google Scholar]
- 218.Yamamoto R, Aoki T, Koseki H, et al. A sphingosine-1-phosphate receptor type 1 agonist, ASP4058, suppresses intracranial aneurysm through promoting endothelial integrity and blocking macrophage transmigration. Br J Pharmacol 2017; 174: 2085–2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Yanagida K, Liu CH, Faraco G, et al. Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci U S A 2017; 114: 4531–4536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Blankenbach KV, Schwalm S, Pfeilschifter J, et al. Sphingosine-1-phosphate receptor-2 antagonists: therapeutic potential and potential risks. Front Pharmacol 2016; 7: 167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Davis PB, Yasothan U, Kirkpatrick P. Ivacaftor. Nat Rev Drug Discov 2012; 11: 349–350. [DOI] [PubMed] [Google Scholar]
- 222.Boyle MP, Bell SC, Konstan MW, et al. A CFTR corrector (lumacaftor) and a CFTR potentiator (ivacaftor) for treatment of patients with cystic fibrosis who have a phe508del CFTR mutation: a phase 2 randomised controlled trial. Lancet Respir Med 2014; 2: 527–538. [DOI] [PubMed] [Google Scholar]
- 223.Guggino WB, Stanton BA. New insights into cystic fibrosis: molecular switches that regulate CFTR. Nat Rev Mol Cell Biol 2006; 7: 426–436. [DOI] [PubMed] [Google Scholar]
- 224.Tsai MF. CFTR: an ion channel with a transporter-type energy-coupling mechanism. J Gen Physiol 2012; 140: 343–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Bulley S, Jaggar JH. Cl− channels in smooth muscle cells. Pflugers Arch 2014; 466: 861–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Eckford PD, Li C, Ramjeesingh M, et al. Cystic fibrosis transmembrane conductance regulator (CFTR) potentiator VX-770 (ivacaftor) opens the defective channel gate of mutant CFTR in a phosphorylation-dependent but ATP-independent manner. J Biol Chem 2012; 287: 36639–36649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Moskwa P, Lorentzen D, Excoffon KJ, et al. A novel host defense system of airways is defective in cystic fibrosis. Am J Respir Crit Care Med 2007; 175: 174–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Berdiev BK, Qadri YJ, Benos DJ. Assessment of the CFTR and ENaC association. Mol Biosyst 2009; 5: 123–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Dudez T, Borot F, Huang S, et al. CFTR in a lipid raft-TNFR1 complex modulates gap junctional intercellular communication and IL-8 secretion. Biochim Biophys Acta 2008; 1783: 779–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Boujaoude LC, Bradshaw-Wilder C, Mao C, et al. Cystic fibrosis transmembrane regulator regulates uptake of sphingoid base phosphates and lysophosphatidic acid: modulation of cellular activity of sphingosine 1-phosphate. J Biol Chem 2001; 276: 35258–35264. [DOI] [PubMed] [Google Scholar]
- 231.Peters W, Kusche-Vihrog K, Oberleithner H, et al. Cystic fibrosis transmembrane conductance regulator is involved in polyphenol-induced swelling of the endothelial glycocalyx. Nanomedicine 2015; 11: 1521–1530. [DOI] [PubMed] [Google Scholar]
- 232.Tousson A, Van Tine BA, Naren AP, et al. Characterization of CFTR expression and chloride channel activity in human endothelia. Am J Physiol 1998; 275: C1555–C1564. [DOI] [PubMed] [Google Scholar]
- 233.Murphy BA, Fakira KA, Song Z, et al. AMP-activated protein kinase and nitric oxide regulate the glucose sensitivity of ventromedial hypothalamic glucose-inhibited neurons. Am J Physiol Cell Physiol 2009; 297: C750–C758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Guo Y, Su M, McNutt MA, et al. Expression and distribution of cystic fibrosis transmembrane conductance regulator in neurons of the human brain. J Histochem Cytochem 2009; 57: 1113–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Bradbury NA, Cohn JA, Venglarik CJ, et al. Biochemical and biophysical identification of cystic fibrosis transmembrane conductance regulator chloride channels as components of endocytic clathrin-coated vesicles. J Biol Chem 1994; 269: 8296–8302. [PubMed] [Google Scholar]
- 236.Ostroumov K, Grandolfo M, Nistri A. The effects induced by the sulphonylurea glibenclamide on the neonatal rat spinal cord indicate a novel mechanism to control neuronal excitability and inhibitory neurotransmission. Br J Pharmacol 2007; 150: 47–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Becq F, Mall MA, Sheppard DN, et al. Pharmacological therapy for cystic fibrosis: from bench to bedside. J Cyst Fibros 2011; 10: S129–S145. [DOI] [PubMed] [Google Scholar]
- 238.Van Goor F, Hadida S, Grootenhuis PD, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci U S A 2009; 106: 18825–18830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Flume PA, Liou TG, Borowitz DS, et al. Ivacaftor in subjects with cystic fibrosis who are homozygous for the F508del-CFTR mutation. Chest 2012; 142: 718–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Cai ZW, Liu J, Li HY, et al. Targeting F508del-CFTR to develop rational new therapies for cystic fibrosis. Acta Pharmacol Sin 2011; 32: 693–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Balch WE, Morimoto RI, Dillin A, et al. Adapting proteostasis for disease intervention. Science 2008; 319: 916–919. [DOI] [PubMed] [Google Scholar]
- 242.Wainwright CE, Elborn JS, Ramsey BW, et al. Lumacaftor-Ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N Engl J Med 2015; 373: 220–231. [DOI] [PubMed] [Google Scholar]
- 243.Sinha C, Zhang W, Moon CS, et al. Capturing the direct binding of CFTR correctors to CFTR by using click chemistry. Chembiochem 2015; 16: 2017–2022. [DOI] [PMC free article] [PubMed] [Google Scholar]