Previous Article in Journal
Decoding Anticancer Drug Response: Comparison of Data-Driven and Pathway-Guided Prediction Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bidirectional Regulation of Nitric Oxide and Endothelin-1 in Cerebral Vasospasm: Mechanisms and Therapeutic Perspectives

1
Institute for Translational Neurosurgery, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
2
Department of Cardiology, Pulmonary Diseases and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
3
Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Future Pharmacol. 2025, 5(4), 59; https://doi.org/10.3390/futurepharmacol5040059 (registering DOI)
Submission received: 15 August 2025 / Revised: 30 September 2025 / Accepted: 7 October 2025 / Published: 10 October 2025

Abstract

Cerebral vasospasm (CVS) following a subarachnoid hemorrhage (SAH) is a critical complication driven by imbalances between vasodilators and vasoconstrictors. This review explores the bidirectional interplay between nitric oxide (NO) and endothelin-1 (ET-1) in CVS pathogenesis. NO, a potent vasodilator mainly produced by endothelial and neuronal nitric oxide synthase (eNOS/nNOS) under normal physiological conditions, is scavenged early after SAH by hemoglobin derivatives, leading to microcirculatory dysfunction, pericyte constriction, and impaired neurovascular coupling. Conversely, ET-1 exacerbates vasoconstriction by suppressing NO synthesis via ROS-dependent eNOS uncoupling and Rho-kinase activation. The NO/ET-1 axis further influences delayed cerebral ischemia (DCI) through mechanisms like 20-HETE-mediated cGMP suppression and oxidative stress. Emerging therapies—including NO donors, NOS gene therapy, and ET-1 receptor antagonists—aim to restore this balance. Understanding these pathways offers translational potential for mitigating CVS and improving outcomes post-SAH.

1. Introduction

Cerebral vasospasm (CVS), which is the widespread constriction of the cerebral vasculature, represents a devastating complication following a subarachnoid hemorrhage (SAH) that significantly contributes to delayed cerebral ischemia (DCI) and poor neurological outcomes. Clinical CVS, diagnosed based on the manifestation of delayed ischemic neurological deficits or reduced territorial levels of CBF in combination with CVS in angiography [1], is also present in up to 16% of angiographically negative cases [2]. However, those that present with more severe pathophysiology and neurological deficits [3,4] and angiographically visible vasospasm do not necessarily include clinical vasospasm [2]. CVS is biphasic, with a delayed phase after 3–5 days and a maximum on day 7, accompanied by delayed cerebral ischemia [5,6,7,8,9]. Despite decades of research, its pathogenesis remains incompletely elucidated. Several mechanisms contribute to the development of CVS. Blood-derived factors play a central role, including the presence of blood clots [10,11,12], hemoglobin [13,14,15], lysed erythrocytes [16,17], and the release of free iron [18]. These compounds trigger a cascade of oxidative and metabolic processes, such as the oxidation of bilirubin [19], activation of heme oxygenase-1 (HO-1) [19], and oxidative stress [20], with superoxide-induced lipid peroxidation further amplifying vascular injury [14,21]. In parallel, inflammatory pathways are strongly implicated [22,23,24,25,26]. Structural and cellular changes also contribute, including the stretching of the arachnoid membrane [27], abnormal contraction of smooth muscle cells, perivascular neuron cell death, remodeling of the arterial wall [9,28,29], and apoptosis [30,31,32]. A variety of messenger molecules and signaling mediators further exacerbate CVS. These include serotonin (5-HT) and endothelin-1 (ET-1) [33,34], prostaglandins [34,35], and sympathetic activation through catecholamines and neuropeptide Y [9,27,34,35,36], as well as other components of the vascular innervation [9,14,37]. Finally, several molecular and lipid signaling pathways have been implicated, such as calcium channel activation in smooth muscle cells [37], sphingolipids (probably derived from thrombocytes) [22], thromboxane [38], MAPK and ERK1/2 signaling [39], and the statin-mediated P3K/Akt/eNOS pathway [40].
Two predominant mechanistic hypotheses dominate the current understanding: nitric oxide (NO) dysregulation and ET-1 overactivation or intracellular signal transduction [41]. Briefly, for the NO hypothesis, early NO depletion due to scavenging by the hemoglobin present in the subarachnoid space after SAH impairs endogenous vasodilation, triggering microcirculatory failure. This loss disrupts the sGC-cGMP (soluble guanylate cyclase and cyclic guanosine monophosphate) pathway and 20-hydroxyeicosatetraenoic acid (20-HETE)-mediated calcium signaling, leading to sustained pericyte constriction and capillary flow arrest. For the ET-1 overactivation hypothesis, ET-1, a potent vasoconstrictor, is upregulated post-SAH through transcriptional activation and mitogen-activated protein kinase (MAPK) phosphorylation, both depending on reactive oxygen species (ROS) as another mediator derived from the hemorrhage in the subarachnoid space. It can directly antagonize NO by suppressing eNOS activity via Rho-kinase (ROCK), while concurrently promoting vascular smooth muscle cell (VSMC) hypercontractility. Notably, although endogenous vasodilation is widely recognized as essential to vascular homeostasis and angiogenesis [41], currently the putative importance of the NO/ET-1 axis and the modulatory roles of intracellular signaling pathways (particularly MAPK in ET-1 receptor trafficking and phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) in NOS phosphorylation) are still not fully emphasized [41].
This review synthesizes advances in the bidirectional NO/ET-1 interplay in cerebral vasospasm pathogenesis, focusing on three critical dimensions: (1) the molecular drivers of axis disruption, including ROS-mediated eNOS uncoupling, 20-HETE-dependent suppression of NO-cGMP signaling, and underemphasized intracellular pathways (MAPK/PI3K-Akt) regulating ET-1 trafficking and NOS phosphorylation; (2) microcirculatory sequelae, spanning pericyte α-SMA (alpha smooth muscle actin) transformation, capillary flow arrest, and cortical spreading depolarizations (CSDs) that amplify delayed cerebral ischemia (DCI); and (3) emerging pharmacotherapies targeting axis recalibration, from hypoxia-activated NO donors and ET-1 antagonists (e.g., clazosentan) to dual-path agents like 20-HETE inhibitors (HET0016).

2. NO-Mediated Vasoregulation: Pharmacological Foundations

NO, a critical endothelium-derived relaxing factor, maintains vascular homeostasis by counteracting vasoconstrictors (e.g., angiotensin II, endothelin-1, 5-HT). Its inhibition triggers vasoconstriction and pressor responses [42,43,44,45], as evidenced by the 50–75% reductions in angiotensine II-induced vasoconstriction upon 20-HETE synthesis inhibition, and the deletion of eNOS in mice in different cell types, which goes hand in hand with hypertension. NO mediates carbon dioxide (CO2)-dependent vasodilation via the canonical sGC/cGMP pathway [12].
Following the classical view, the vasodilatory response to NO is secondary to the activation of sGC and a subsequent elevation of cGMP [12,44]. NO binding to sGC elevates cGMP, activating calcium-activated K+ (KCa) channels to sequester cytosolic Ca2+ into the sarcoplasmic reticulum, thereby relaxing vascular smooth muscle cells [13,46] and acting as a pericyte dilator [14]. This mechanism underpins CO2-induced vasodilation in cerebral vessels.
However, more recent investigations have shown the importance of the cGMP-independent modulation of NO levels, e.g., through 20-HETE [13,42,44]. This cytochrome P450 (CYP4A)-derived metabolite suppresses NO bioactivity by blocking KCa channels, causing VSMC depolarization and impaired normal SMC relaxation [14,44]. It also modulates GC via P/Q type channels and N-methyl-D-aspartate (NMDA) receptor modulation [47]. Intriguingly, NO reciprocally inhibits 20-HETE synthesis by binding to the heme moiety of CYP4A enzymes—a critical regulatory node where diverse vasoconstrictive pathways (e.g., PKC/Rho-kinase) converge to amplify 20-HETE production [13,44].
The spatiotemporal dynamics of NO signaling are governed by three nitric oxide synthase (NOS) isoforms with distinct pharmacological profiles [18,48]. Under physiological conditions, endothelial NOS (eNOS/NOS3), activated by shear stress or PI3K-Akt phosphorylation, generates NO for sustained vasodilation and pericyte relaxation, serving as the primary regulator of basal vascular tone and vasodilation [15,18,48,49,50,51]. However, some studies reported that, in SAH, oxidative stress can lead to “eNOS uncoupling,” where the enzyme produces a superoxide instead of NO, thereby exacerbating vascular dysfunction and oxidative injury [52]. Neuronal NOS, formerly named non-inducible NOS (nNOS/NOS1), localized in perivascular neurons and pericytes [14,50], is a constitutive enzyme that mediates neurovascular coupling and central blood pressure regulation. While numerous reports indicate nNOS-derived NO can upregulate cytoprotective antioxidant defenses, some reported that its hyperactivity under a few pathological conditions may contribute to excitotoxic damage [53]. Notably, both eNOS and nNOS are constitutive enzymes, but their activity can be enhanced by various stimuli. Conversely, inducible NOS (iNOS/NOS2) is expressed at low levels under physiological conditions, but is robustly upregulated during inflammation in a wide range of cells post-SAH, including macrophages, microglia, vascular smooth muscle cells, and pericytes [52,54]. The role of iNOS is profoundly dual and context-dependent. On one hand, its high-output NO production can be pathogenic, generating peroxynitrite and driving cerebral vasospasm (CVS) and neuronal apoptosis [52]. On the other hand, iNOS also possesses critical regulatory functions, such as suppressing pathogenic T-cells and promoting the infiltration of regulatory myeloid cells, which can help control neuroinflammation [55].
Under physiological conditions, endothelial-derived NO diffuses to adjacent SMCs (<100 μm), directly mediating VSMC relaxation [18], while neuronal NO (from nNOS) fine-tunes microvascular tone through perivascular nerve plexuses that anatomically bridge nitrogenergic neurons and cerebral microvessels. Notably, during neuronal activity-regulated vasodilation, the close proximity between capillaries and neurons (<50 μm) enables efficient NO signaling despite its limited diffusion range (<200 μm) [47]. However, in pathological contexts such as ischemia–reperfusion (I/R), microvessels become the dominant NO source due to endothelial activation and inflammatory cell infiltration [15]. This shift is functionally critical, as I/R-induced oxidative stress rapidly scavenges NO, exacerbating microcirculatory dysfunction despite increased production.
Pharmacologically, this multifaceted regulation offers actionable targets; in particular, the interplay between NOS isoforms, 20-HETE, and downstream effectors thus defines a druggable axis for vascular pathologies.

3. NO Dysregulation Post-SAH: Therapeutic Challenges

NO is critically involved in regulating cerebral blood flow (CBF) after SAH, with its depletion driving disturbances in the microcirculation. These include capillary flow impairment, microvascular constriction at pericyte sites, and reduced mean transit time (MTT), which is closely correlated with decreased NO levels and increased capillary transit time heterogeneity (CTH), alongside large vessel cerebral vasospasm (CVS) [12,14,15,27,44,56,57,58,59]. Concurrently, CO2 reactivity diminishes [12], yet Cseplo et al. observed a preserved CO2 reactivity in large vessels despite hemoglobin-induced NO scavenging [46].
NO levels decrease during the early phase post-SAH in both human and animal models [60,61], consistent with its known reduction under inflammatory conditions in general [62]. This depletion post-SAH occurs primarily because extravasated blood scavenges NO via oxygenated hemoglobin (oxyHb), bilirubin, and iron [15,25,44,63,64], resulting in a transient fall in CBF. Endogenous NOS inhibitors and the degeneration of perivascular nitrergic neurons further reduce early NO availability [14,15,44,65,66]. For example, asymmetric dimethyl arginine (ADMA), of which the degradation is inhibited by hemoglobin metabolism (bilirubin-oxidized fragments), inhibits the physiological upregulation of eNOS upon vasoconstriction-mediated shear stress. Notably, while the upregulation of NO products in the brain appears to oppose the sustained vasoconstrictor response to hemoglobin, these factors (ADMA accumulation and neuronal degeneration) act as causatives for the prolonged fall in CBF (30 min until 12 h). Concurrently, tissue hypoxia exacerbates this loss by limiting the oxygen substrate required for NO synthesis [14]. This initiates a vicious cycle: NO depletion leads to hypoxia, which further aggravates NO deficiency. Furthermore, ROS play a central role in the loss of NO bioavailability after SAH. Extravasated blood in the subarachnoid space generates a significant oxidative burden, for instance, through the autoxidation of oxyhemoglobin. Among these ROS, the superoxide anion (O2) reacts with NO at an extremely rapid rate, forming the potent oxidant peroxynitrite (ONOO) [67]. This reaction not only directly scavenges vasoprotective NO but also yields a product that inflicts further damage. Peroxynitrite contributes to a vicious cycle by inducing eNOS uncoupling, likely through the oxidation of the essential eNOS cofactor tetrahydrobiopterin (BH4) [67]. This converts eNOS from a protective NO synthase into a source of additional superoxide, drastically amplifying oxidative stress and depleting NO. Concurrently, the endogenous antioxidant defense systems, including enzymes like superoxide dismutase and the glutathione system, become overwhelmed and deficient [68]. This failure of antioxidant capacity allows ROS to accumulate unchecked, accelerating NO consumption and directly compromising endothelial function. However, in contradiction to observations implicating NOS reduction in prolonged CBF decline, some studies reported no effect of NOS inhibition on CBF [15,44]. In the longer run (12–24 h post-SAH), NO metabolites (nitrite/nitrate) and eNOS increase in the CSF [69], suggesting compensatory mechanisms despite persistent microcirculatory dysfunction.
By day 7 post-SAH, oxygenated hemoglobin (oxyHb) from hemolysis peaks, coinciding with the maximal CVS severity, induces vasoconstriction through the ROS generated during its autoxidation to methemoglobin (metHb) [14,44,69]. Heme degradation by heme oxygenase-1 (HO-1) releases bilirubin, carbon monoxide (CO), and iron, fueling peroxynitrite formation and elevated nitrotyrosine [44]. Peroxynitrite itself acts as a vasoconstrictor by impairing smooth muscle cell relaxation [14,69], inactivating tissue plasminogen activator (tPA) to increase thrombogenicity, and causing tissue damage via lipid peroxidation, poly(ADP-ribose)-polymerase (PARP) activation, and mitochondrial dysfunction [14,70], thereby worsening the post-SAH energy crisis and amplifying ROS production.
Mechanistically, NO depletion inhibits the NO-cGMP pathway, triggering pericyte α-SMA transformation and constriction [14,15]. Oxyhemoglobin also mediates vasoconstriction by altering SMC potassium channels, upregulating voltage-dependent calcium channels, and modulating transient receptor potential channels [63]. Notably, hemoglobin-induced NO scavenging partly stems from increased 20-HETE production; blocking 20-HETE prevents CBF reduction after blood/Hb injection and normalizes vascular responses to NOS inhibition [44]. Similar effects can also be observed after cerebral ischemia [15].
Other reasons for the microcirculatory failure may be due to NO’s role in suppressing thrombocyte and neutrophil activation and vascular adhesion [27]—low NO levels correlate with microthrombosis extents [14]. Reduced NO (alongside elevated K+) also lowers the threshold for CSDs. These propagate via P/Q-type calcium channels and NMDA receptor modulation [47,71], potentially driving the inverse haemodynamic response and spreading ischemia after SAH [65,71]. Repetitive CSDs exacerbate microvascular spasms and NO resistance through intracellular Ca2+ accumulation or matrix metalloproteinase-9 (MMP-9) activation, mechanistically explaining delayed ischemic neurological deficits (DINDs) [72,73].
Beyond CBF dysregulation, NO contributes to tissue damage [72,73]. While crucial for vasodilation, its overproduction, particularly in reaction with superoxide to form peroxynitrite (ONOO), can mediate glutamate excitotoxicity and oxidative stress, leading to neuronal death [74,75]. This pathway is implicated in inducing mitochondrial dysfunction and apoptotic signaling [75]. Concurrently, elevated NO can disrupt the blood–brain barrier (BBB), potentially by mediating VEGF-induced increases in permeability [44,75]. Endothelial dysfunction is another component of the tissue damage after SAH, inducing an imbalance in vessel tone regulation [76]. An overproduction of NO via inflammatory induction contributes to cytotoxic and structural damage [14,77].
Paradoxically, NO reduction in the microcirculation links to delayed cerebral ischemia [47,57,78], and NO depletion leads to CSD promotion and cortical spreading ischemia (CSI), which further contribute to the development of DCI and DIND [47,72,79,80], while NO donors confer neuroprotection by sustaining microvascular oxygen delivery during the critical phase post-SAH or ischemia–reperfusion [14].
NOS isoform polymorphisms and dynamics further complicate the pathology and the relevant therapy [81]. nNOS (constitutively cytoprotective) disappears from neuronal fibers in the arterial adventitia after SAH [14], due to hemoglobin-induced neuronal destruction [15]. ET-1 also inhibits nNOS-derived NO, impairing functional hyperemia and neurovascular regulation [82]. eNOS, a major endothelial source of NO, becomes phosphorylated through ROS-induced peroxynitrite-mediated uncoupling after SAH, resulting in reduced NO levels [83]. The main source of NO after brain I/R is the microvessels/capillaries [14,15], with reduced levels of eNOS after SAH [14,80], for which microcirculation-related damages are one probable cause [15], though also an increased production of NO due to an increase in eNOS in the walls of pial arteries has been observed in the first days after SAH, which is followed by eNOS reduction and accompanied by an increase in NO breakdown products [14]. eNOS is also found in pericytes under inflammatory conditions in SAH [15,84], negatively affecting macrophage infiltration in experimental treatments of cerebral aneurysm rupture [84].The effects of eNOS phosphorylation/activation after SAH as well as NO production are still controversial; they cause neuroprotection and injury [15,40]. Due to its vasodilatory effects, eNOS exerts a protective effect in the early stages of ischemia [14,18]. Its dysfunction contributes to the impairment of vasodilation upon SAH [62], rendering it a contributor to the development of CVS [40,44,69,85], e.g., via promoting an increased adherence and infiltration of leukocytes in cerebral arteries [14,80], via p53 inhibiting SMC proliferation [30] and via promoting the alpha-SMA transformation of pericytes, which causes microvessel constriction [15]. Disturbances in the microcirculation are also promoted by increased levels of NO. It also contributes to preserving CBF autoregulation in rat pial arteries in the acute stage after SAH, probably via the scavenging of superoxide anions by NO (lower limit increased, eNOS and superoxide anion increased in hypotension, L-arginine preserved) [86], and reduces the development of neurological deficits after SAH [15]. In contrast to this, the inducible, proinflammatory NOS isoform, iNOS, represents a pivotal and complex player in SAH pathophysiology. Under physiological conditions, iNOS is barely detectable in cerebral vessels [85]. However, following SAH, it is found at increased levels in macrophages and mononuclear and polymorphonuclear cells in the subarachnoid space. In cerebral vessel walls, it is present in SMCs, endothelial cells, and adventitial cells, and under the inflammatory conditions in SAH can furthermore be found in microglia, neurons, leukocytes, thrombocytes, fibroblasts, and pericytes [15,18,48,62,70,84,87]. This expression is driven by a complex inflammatory cascade involving cytokines and master regulators like NF-κB, which is itself activated by ROS and PARP [18,26,48,58,64,87,88]. As previously mentioned, the role of iNOS in pathophysiology is profoundly dual and context-dependent. On one hand, its high-output, sustained production of NO is a well-established pathogenic mechanism. On the other hand, a more nuanced understanding has emerged, revealing that iNOS also possesses critical immunoregulatory functions. This duality explains why a global inhibition of iNOS has shown mixed outcomes, sometimes worsening pathology [55]. Therefore, the therapeutic paradigm is shifting. While iNOS remains a high-value target due to its clear detrimental effects, the strategy is evolving from one of simple blockade to one of fine-tuning, for instance, by ensuring proper cofactor availability to prevent uncoupling.

4. ET-1/NO Antagonism: Pharmacodynamic Interplay

Despite conclusive evidence of the NO/ET-1 axis critically regulating vascular homeostasis [85,89], its therapeutic exploitation remains underdeveloped. The bidirectional imbalance between NO and ET-1 constitutes a convergent pathway in cerebral vasospasm pathogenesis, operating through reciprocal molecular suppression (Figure 1). When ET-1 binds ETA receptors, it activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase to generate superoxide (O2), which scavenges NO to form peroxynitrite (ONOO)—a process that oxidizes eNOS cofactors, converting this critical enzyme from an NO synthase to an ROS generator (uncoupling), while increasing nitrotyrosine levels [18,69]. This ET-1-mediated eNOS inactivation occurs through ROCK-dependent phosphorylation changes, simultaneously suppressing neuronal NO production by nNOS and impairing activity-dependent vasodilation (neurovascular uncoupling) [15,82]. This dual inhibition is particularly consequential after SAH, where nNOS disappears from neuronal fibers in the arterial adventitia and nNOS-containing neurons are destroyed by hemoglobin [14]. Compounding this damage, ET-1 inhibits NO production by the surviving nNOS, reducing functional hyperemia and impairing neurovascular regulation [82,90].
The resulting oxidative stress manifests through multiple pathways: ET-1 induces mitochondrial ROS, eNOS uncoupling, and the activation of NADPH oxidase in vascular cells [82]. These effects render the brain more susceptible to injury by compromising the balance between energy demands and blood flow delivery. Crucially, hemoglobin breakdown products (methaemoglobin [metHb], bilirubin) disrupt the ET-1/NO equilibrium by simultaneously scavenging NO and amplifying ET-1 release, establishing a self-perpetuating cycle of vasoconstriction. This imbalance extends beyond macrovascular spasms to drive microcirculatory failure, which is manifested through a pericyte α-SMA transformation and capillary stall via 20-HETE accumulation and ET-1-induced RhoA/ROCK activation.
Conversely, physiological NO suppresses ET-1 transcription through counter-regulatory suppression. It activates the cGMP-PKG (protein kinase G)-mediated inhibition of nuclear factor kappa B (NF-κB) and activating protein-1 (AP-1) signaling cascades, reducing ET-1 gene expression while downregulating vasoconstrictive ETA receptors and preserving protective ETB receptors that mediate NO/prostacyclin release. Nerve fiber stimulation exploits this pathway, with NO release relieving angiographic vasospasm. The PI3K/Akt pathway mediates ischemic tolerance by maintaining endothelial cell survival and NO-mediated vascular tone regulation, resulting in reduced vasospasm. This protective mechanism involves eNOS phosphorylation through PI3K/Akt activation (e.g., by statins) [15,40,85] and the DCC (deleted in colorectal cancer)-ERK1/2 (extracellular signal-regulated kinase)-eNOS-NO feed-forward loop after cardiac I/R [15], which counteracts ET-1’s damaging effects.
Pathologically, SAH triggers a vicious cycle initiated by hemoglobin breakdown products, oxyHb and bilirubin, that simultaneously scavenge NO and amplify ET-1 secretion. This dual assault drives microcirculatory collapse through ET-1-induced pericyte α-SMA transformation (via RhoA/ROCK) and concurrent NO depletion blocking KCa channels, preventing VSMC hyperpolarization. Oxidative escalation follows as ET-1-activated NADPH oxidase generates O2 that fuels mitochondrial ROS production, converting residual NO to vasoconstrictive ONOO while inducing eNOS uncoupling for further NO loss. Secondary injury cascades then manifest: neurovascular decoupling from nNOS degeneration lowers the threshold for cortical spreading depolarizations; ET-1-induced vasoconstriction converts these into “spreading ischemia”; and thromboinflammation erupts from lost NO’s antiplatelet effects combined with ET-1’s P-selectin induction. The resulting hypoxia perpetuates this cycle, rendering the brain increasingly vulnerable to energy supply–demand mismatch.
This self-reinforcing pathophysiology extends beyond macrovascular spasms to encompass capillary stalls, neutrophil plugging, and expanding infarct cores, all rooted in the ET-1/NO axis disruption where each molecule’s dysfunction amplifies the other’s downstream damage.

5. Pharmacological Interventions: Targeting the NO/ET-1 Axis

Pharmacological strategies targeting the NO/ET-1 axis restore vasodilatory–vasoconstrictive balance, inhibit oxidative stress/inflammation, and reverse vascular remodeling, offering core therapeutic approaches for cardiovascular/cerebrovascular and renal diseases. As shown in Table 1, the current drug development focuses on the following strategies.
For the restoration of the vasodilatory signaling, NO donors and analogs were widely considered. Classic nitrates like sodium nitroprusside could directly release NO, activating the sGC-cGMP pathway for rapid vascular smooth muscle relaxation. Clinical studies confirm that intrathecal administration significantly alleviates CVS post-SAH. Recently, successful therapeutic strategies against CVS have been applied using NO donors (L-Arginine, nitrite) and NO synthase gene therapy [13,14,80,101,102,103]. But, also, early NO donor treatment has been observed to increase CBF, e.g., due to preventing CTH increases, while, due to a hypotensive side effect, blood pressure and CPP are reduced, which are potentially beneficial effects due to hypertension often being a comorbidity in the presence of SAH [14,57,58,59]. Endogenous nitrite is converted to NO in the tissue without the need for oxygen as a substrate; thus, upon the infusion of nitrite, NO depletion and ROS production are reduced [14]. In addition, nitrite reduces mitochondrial proton leakage, increasing ATP yields from oxygen and thereby tissue tolerance to oxygen reduction [14], and may attenuate thrombogenicity by inhibiting thrombocyte aggregation [14]. Scientists have also tried to develop next-generation NO donors such as benzodifuroxan derivatives which can release NO via non-enzymatic pathways, generating reactive nitrogen species (e.g., NO) that efficiently activate sGC even in hypoxic tissues, thus overcoming drug resistance [104,105]. Inorganic nitrites like sodium nitrite could also be converted to NO by tissue reductases without oxygen dependence, enhancing hypoxic tolerance. SAH models show 15–30% increased CBF and reduced mitochondrial proton leakage to boost ATP synthesis. Still, systemic hypotension (>25% incidence) and tachyphylaxis (50% efficacy loss after chronic use) remain key challenges in the clinical trials.
ET-1 receptor antagonists were also considered as an effective pharmacological intervention approach, as they can efficiently block vasoconstrictive signaling [106]. ETA antagonists such as Ambrisentan can specifically block ETA receptors, inhibiting ET-1-mediated vasoconstriction/smooth muscle proliferation. It has already been used for pulmonary arterial hypertension (PAH) but carries peripheral edema risk (28.6% incidence) [92]. Clazosentan could reduce CVS incidence by 43% in SAH Phase II trials but failed to significantly improve delayed cerebral ischemia outcomes [91]. Dual ETA/ETB antagonists were also considered helpful in this approach. For example, Macitentan/Sparsentan can block ETA (inhibit constriction), while partially activating ETB (promoting NO release) [107]. Sparsentan has been shown to be able to reduce the urine albumin–creatinine ratio (UACR) by 49.5% in diabetic nephropathy trials vs. 33.1% with angiotensin receptor blockers (ARBs) [108]. Advanced delivery systems were also developed to produce a sustainable supply of the ET-1 receptor antagonists. For example, the PER-001 intravitreal implant, which can make a sustained release of ETA antagonist within a 6-month duration, has been applied in a diabetic retinopathy Phase 2a trial. Results from this trial indicate that the treatment significantly reduced macular ischemia and improved low-luminance visual acuity [109].
Beyond directly supplementing NO or blocking ET-1 receptors, a complementary therapeutic strategy aims to enhance endogenous NO bioavailability by targeting the underlying oxidative stress. In this context, thiol-based antioxidants such as N-acetylcysteine (NAC) and glutathione present a promising approach [93]. These agents do not directly donate NO but instead act as potent scavengers of ROS, thereby shielding NO from premature degradation and potentially reversing eNOS uncoupling. This restoration of the redox balance helps to preserve the integrity of the NO-sGC-cGMP signaling pathway, effectively counteracting ET-1-induced vasoconstriction and endothelial dysfunction from a different nodal point. This strategy is particularly relevant in the highly oxidative environment of SAH, where the efficacy of conventional agents like L-arginine can be compromised.
Pharmacological targeting of the NO/ET-1 axis is evolving from single-pathway modulation to multi-pathway coordination. As shown in Table 2, over the recent decades, scientists have also focused on the development of multi-target agents modulating NO/ET-1 balance, each with a unique mechanism to restore the NO-cGMP signaling. A prime example is the 20-HETE synthesis inhibitor TS-011 [44]. By blocking the production of 20-HETE, it not only prevents hemoglobin-induced vasospasm but, more importantly, demonstrates the capability to reverse established vasospasm when administered several days post-SAH, normalizing the vascular diameter [44]. Furthermore, by antagonizing specific G protein-coupled receptors for 20-HETE such as GPR75, significant drivers of hypertension and vascular dysfunction, such as the CYP/20-HETE/GPR75 axis, could be effectively suppressed [110]. Thus, this approach presents a promising strategy to block the deleterious effects of 20-HETE, including vascular inflammation, endothelial dysfunction, and the sensitization to constrictor stimuli, without affecting its synthesis, offering a novel means to rebalance the NO/ET-1 axis and mitigate cerebrovascular diseases.
Of note, herbal active components such as Tetramethylpyrazine (TMP) from Ligusticum chuanxiong have demonstrated the ability to suppress cerebral vasospasm by upregulating eNOS activity and inhibiting ET-1 overproduction, synergizing with nimodipine to improve cerebral blood flow in SAH models [116,117]. Besides these approaches, innovative combination strategies and delivery systems were also investigated to further improve the therapeutic potential. Sequential combination therapy, such as the PAH protocol, combines ERA (e.g., Macitentan) and phosphodiesterase (PDE)-5i (e.g., Sildenafil) for the low-risk group, while using Prostacyclin for the high-risk group, significantly improving the 5-year survival rate to over 75% [118]. Researchers also focused on the SAH window; early nitrite (anti-hypoxia) and delayed ETA antagonist (anti-remodeling) were selectively suggested to avoid hypotension [119,120,121]. Nanodelivery systems such as the liposome-encapsulated eNOS gene or eNOS-activating nanoparticles targeting cerebrovascular endothelia have been considered to increase local NO production [100]. Personalized therapies which stratify patients by their individual ET-1/NO ratio or ADMA (endogenous NOS inhibitor) levels might be recommended for precision dosing [122]. Emerging targets in this pharmacological field also include sGC allosteric activators such as BAY 58-2667, which can activate oxidized sGC and is effective in high-oxidative SAH conditions [123], as well as ETB-biased agonists which can selectively stimulate the ETB-NO pathway without ETA-mediated side effects [124].
Yet, for further drug development, certain existing pharmacokinetic barriers still limit the clinical translation outcome. For example, a low BBB penetration limits many ET-1 antagonists from reaching the central nervous system (CNS), limiting CVS/DCI efficacy. The short NO donor half-life also demands more sustained-release formulations. Future breakthroughs will rely on innovative drug design (e.g., long-acting NO donors), precision delivery (e.g., brain/ocular implants), and phenotype-guided combinations to achieve comprehensive benefits—from vasospasm relief to end-organ protection.

6. Discussion

The emerging evidence suggests that the bidirectional regulation of NO and ET-1 represents a central pathogenic axis in the development of CVS and DCI following SAH. As reviewed, the imbalance between vasodilatory NO and vasoconstrictive ET-1 drives microcirculatory dysfunction, pericyte constriction, oxidative stress, and neurovascular uncoupling. Pharmacological strategies aimed at restoring this balance such as NO donors, ET-1 receptor antagonists, and multi-target agents like 20-HETE inhibitors have shown promise in preclinical and early clinical studies. However, the translational success of these approaches has been limited by challenges such as systemic hypotension, poor blood–brain barrier penetration, and the complexity of NO signaling dynamics in the ischemic brain.
A critical layer of complexity arises from the dual pathways of NO generation. Beyond the canonical NOS-dependent pathway [125], the NOS-independent nitrate–nitrite–NO pathway serves as a vital backup system, particularly under hypoxic conditions prevalent in SAH [126]. While the canonical pathway is compromised by ET-1-mediated inhibition, ROS-induced eNOS uncoupling, and oxygen scarcity, the reduction of nitrite to NO by deoxyhemoglobin and other 5-coordinated ferrous hemes (e.g., in cytochromes) is enhanced in hypoxia [127]. Importantly, the substrate nitrite is readily available from plasma and brain tissue, and can be replenished from dietary nitrate via microbial reduction in the oral cavity and gut, establishing a clinically accessible “enterosalivary nitrate-nitrite-NO” axis [128]. Under the specific SAH pathophysiological conditions, leveraging the nitrite–NO pathway represents a strategic bypass to restore NO bioavailability despite high ET-1 tones. This explains the efficacy of sodium nitrite in experimental SAH, improving CBF without relying on compromised NOS activity, probably also attenuating mitochondrial dysfunction [14,129,130,131]. This NOS-independent pathway may serve as a critical backup mechanism to maintain NO bioavailability when the canonical pathway is impaired, offering a promising therapeutic avenue for conditions like SAH where hypoxia and NOS dysfunction coexist.
Therefore, future therapeutic strategies should not only target the NO-ET-1 axis via conventional NOS modulation or ET-1 antagonism but also exploit the nitrite–NO pathway to bypass NOS limitations for the optimal outcomes. Interventions such as nitrite infusion, hypoxia-activated NO donors, or agents that enhance nitrite reduction could provide sustained NO delivery without exacerbating oxidative stress or depending on oxygen availability.

7. Conclusions

The NO-ET-1 axis plays a pivotal role in the pathophysiology of cerebral vasospasm and delayed cerebral ischemia after SAH. While the current therapeutic approaches focus on rebalancing this axis through NO donors, ET-1 antagonists, and multi-target agents, their clinical efficacy remains partial. A deeper understanding of NO biology offers new insights and opportunities for intervention. By integrating strategies that enhance both canonical and alternative NO sources, and by timing interventions to align with the evolving pathophysiology of SAH, we may better address the multifactorial nature of CVS and DCI. Future research should prioritize the development of targeted, hypoxia-sensitive NO delivery systems and personalized treatment regimens based on individual NO/ET-1 profiles and redox status, ultimately improving outcomes for patients suffering from this devastating condition.

Author Contributions

Conceptualization, K.B., K.L.; writing—original draft preparation, review and editing, K.B., K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

We would like to thank Heinrich Heine University Duesseldorf and University Hospital Duesseldorf for their support.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
20-HETE20-Hydroxyeicosatetraenoic Acid
α-SMAalpha Smooth Muscle Actin
ARBsAngiotensin Receptor Blockers
ACEiAngiotensin-Converting Enzyme Inhibitors
AngIIAngiotensin II
ADMAAsymmetric Dimethyl Arginine
BBBBlood–Brain Barrier
CBFCerebral Blood Flow
cGMPCyclic Guanosine Monophosphate
CNSCentral Nervous System
CO2Carbon Dioxide
CSDCortical Spreading Depolarizations
CSICortical Spreading Ischemia
CVSCerebral Vasospasm
CTHTransit Time Heterogeneity
CYPA4Cytochrome P450
DCCDeleted in Colorectal Cancer
DCIDelayed Cerebral Ischemia
DINDDelayed Ischemic Neurological Deficit
eNOSEndothelial NOS
ERKExtracellular Signal Regulated Kinase
ET-1Endothelin-1
HO-1Heme Oxygenase-1
iNOSInducible NOS
I/RIschemia/Reperfusion
KCa channelsCalcium-Activated K+ Channels
L-NAMEL-NG-Nitro Arginine Methyl Ester
MAPKMitogen Activated Protein Kinase
metHbMethaemoglobin
MMP9Matrix Metalloproteinase 9
MnSODMangan Superoxide Dismutase
MTTMean Transit Time
NADPHNicotinamide Dinucleotide Phosphate
NF-κBNuclear Factor Kappa B
NMDAN-Methyl-D-Aspartate
NONitric Oxide
NOSNitric Oxide Synthase
nNOSNeuronal NOS
O2Superoxide
ONOOPeroxynitrite
oxyHbOxygenated Hemoglobin
PAHPulmonary Arterial Hypertension
PARPPoly(Adenosine Diphosphate-Ribose)-Polymerase
PDEPhosphodiesterase
PI3K/AktPhosphatidylinositol-3-Kinase/Protein Kinase B
PKGProtein Kinase G
ROCKRho-Associated Protein Kinase
ROSReactive Oxygen Species
SAHSubarachnoid Hemorrhage
sGCSoluble Guanylate Cyclase
TMPTetramethylpyrazine
tPATissue Plasminogen Activator
VEGFVascular Endothelial Growth Factor
VSMCVascular Smooth Muscle Cell

References

  1. Vajkoczy, P.; Horn, P.; Thome, C.; Munch, E.; Schmiedek, P. Regional cerebral blood flow monitoring in the diagnosis of delayed ischemia following aneurysmal subarachnoid hemorrhage. J. Neurosurg. 2003, 98, 1227–1234. [Google Scholar] [CrossRef]
  2. Gross, B.A.; Lin, N.; Frerichs, K.U.; Du, R. Vasospasm after spontaneous angiographically negative subarachnoid hemorrhage. Acta Neurochir. 2012, 154, 1127–1133. [Google Scholar] [CrossRef]
  3. Hui, F.K.; Tumialán, L.M.; Tanaka, T.; Cawley, C.M.; Zhang, Y.J. Clinical Differences Between Angiographically Negative, Diffuse Subarachnoid Hemorrhage and Perimesencephalic Subarachnoid Hemorrhage. Neurocritical Care 2009, 11, 64–70. [Google Scholar] [CrossRef]
  4. Grubb, R.L.; Raichle, M.E.; Eichling, J.O.; Gado, M.H. Effects of subarachnoid hemorrhage on cerebral blood volume, blood flow, and oxygen utilization in humans. J. Neurosurg. 1977, 46, 446–453. [Google Scholar] [CrossRef]
  5. Kurki, M.I.; Häkkinen, S.-K.; Frösen, J.; Tulamo, R.; Von Und Zu Fraunberg, M.; Wong, G.; Tromp, G.; Niemelä, M.; Hernesniemi, J.; Jääskeläinen, J.E.; et al. Upregulated Signaling Pathways in Ruptured Human Saccular Intracranial Aneurysm Wall: An Emerging Regulative Role of Toll-Like Receptor Signaling and Nuclear Factor-κB, Hypoxia-Inducible Factor-1A, and ETS Transcription Factors. Neurosurgery 2011, 68, 1667–1676. [Google Scholar] [CrossRef] [PubMed]
  6. Sehba, F.A.; Hou, J.; Pluta, R.M.; Zhang, J.H. The importance of early brain injury after subarachnoid hemorrhage. Prog. Neurobiol. 2012, 97, 14–37. [Google Scholar] [CrossRef] [PubMed]
  7. Cai, J.; Sun, Y.; Yuan, F.; Chen, L.; He, C.; Bao, Y.; Chen, Z.; Lou, M.; Xia, W.; Yang, G.-Y.; et al. A Novel Intravital Method to Evaluate Cerebral Vasospasm in Rat Models of Subarachnoid Hemorrhage: A Study with Synchrotron Radiation Angiography. PLoS ONE 2012, 7, e33366. [Google Scholar] [CrossRef] [PubMed]
  8. Osaka, K. Prolonged vasospasm produced by the breakdown products of erythrocytes. J. Neurosurg. 1977, 47, 403–411. [Google Scholar] [CrossRef]
  9. Cetas, J.; Lee, D.; Alkayed, N.; Wang, R.; Iliff, J.; Heinricher, M. Brainstem control of cerebral blood flow and application to acute vasospasm following experimental subarachnoid hemorrhage. Neuroscience 2009, 163, 719–729. [Google Scholar] [CrossRef]
  10. Güresir, E.; Schuss, P.; Borger, V.; Vatter, H. Experimental Subarachnoid Hemorrhage: Double Cisterna Magna Injection Rat Model—Assessment of Delayed Pathological Effects of Cerebral Vasospasm. Transl. Stroke Res. 2015, 6, 242–251. [Google Scholar] [CrossRef]
  11. Jakobsen, M.; Overgaard, J.; Marcussen, E.; Enevoldsen, E.M. Relation between angiographic cerebral vasospasm and regional CBF in patients with SAH. Acta Neurol. Scand. 1990, 82, 109–115. [Google Scholar] [CrossRef]
  12. Friedrich, B.; Michalik, R.; Oniszczuk, A.; Abubaker, K.; Kozniewska, E.; Plesnila, N. CO2 Has no Therapeutic Effect on Early Micro Vasospasm after Experimental Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2014, 34, e1–e6. [Google Scholar] [CrossRef]
  13. Takeuchi, K.; Miyata, N.; Renic, M.; Harder, D.R.; Roman, R.J. Hemoglobin, NO, and 20-HETE interactions in mediating cerebral vasoconstriction following SAH. Am. J. Physiol. Integr. Comp. Physiol. 2006, 290, R84–R89. [Google Scholar] [CrossRef] [PubMed]
  14. Østergaard, L.; Aamand, R.; Karabegovic, S.; Tietze, A.; Blicher, J.U.; Mikkelsen, I.K.; Iversen, N.K.; Secher, N.; Engedal, T.S.; Anzabi, M.; et al. The Role of the Microcirculation in Delayed Cerebral Ischemia and Chronic Degenerative Changes after Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2013, 33, 1825–1837. [Google Scholar] [CrossRef]
  15. Li, Q.; Chen, Y.; Li, B.; Luo, C.; Zuo, S.; Liu, X.; Zhang, J.H.; Ruan, H.; Feng, H. Hemoglobin induced NO/cGMP suppression Deteriorate Microcirculation via Pericyte Phenotype Transformation after Subarachnoid Hemorrhage in Rats. Sci. Rep. 2016, 6, 22070. [Google Scholar] [CrossRef] [PubMed]
  16. Greenhalgh, A.D.; Brough, D.; Robinson, E.M.; Girard, S.; Rothwell, N.J.; Allan, S.M. Interleukin-1 receptor antagonist is beneficial after subarachnoid haemorrhage in rat by blocking haem-driven inflammatory pathology. Dis. Model. Mech. 2012, 5, 823–833. [Google Scholar] [CrossRef]
  17. Bradley, W.G.; Schmidt, P.G. Effect of methemoglobin formation on the MR appearance of subarachnoid hemorrhage. Radiology 1985, 156, 99–103. [Google Scholar] [CrossRef]
  18. Chen, L.-C.; Lee, W.-S. Estradiol Reduces Ferrous Citrate Complex-Induced NOS2 Up-Regulation in Cerebral Endothelial Cells by Interfering the Nuclear Factor Kappa B Transactivation through an Estrogen Receptor β–Mediated Pathway. PLoS ONE 2013, 8, e84320. [Google Scholar] [CrossRef] [PubMed]
  19. Sharp, F.R.; Zhan, X.; Liu, D.-Z. Heat Shock Proteins in the Brain: Role of Hsp70, Hsp 27, and HO-1 (Hsp32) and Their Therapeutic Potential. Transl. Stroke Res. 2013, 4, 685–692. [Google Scholar] [CrossRef]
  20. Yang, Y.; Chen, S.; Zhang, J.-M. The Updated Role of Oxidative Stress in Subarachnoid Hemorrhage. Curr. Drug Deliv. 2016, 14, 832–842. [Google Scholar] [CrossRef]
  21. Echigo, R.; Shimohata, N.; Karatsu, K.; Yano, F.; Kayasuga-Kariya, Y.; Fujisawa, A.; Ohto, T.; Kita, Y.; Nakamura, M.; Suzuki, S.; et al. Trehalose treatment suppresses inflammation, oxidative stress, and vasospasm induced by experimental subarachnoid hemorrhage. J. Transl. Med. 2012, 10, 80. [Google Scholar] [CrossRef]
  22. Wirrig, C.; Hunter, I.; Mathieson, F.A.; Nixon, G.F. Sphingosylphosphorylcholine is a Proinflammatory Mediator in Cerebral Arteries. J. Cereb. Blood Flow Metab. 2011, 31, 212–221. [Google Scholar] [CrossRef]
  23. Handa, Y.; Kabuto, M.; Kobayashi, H.; Kawano, H.; Takeuchi, H.; Hayashi, M. The correlation between immunological reaction in the arterial wall and the time course of the development of cerebral vasospasm in a primate model. Neurosurgery 1991, 28, 542–549. [Google Scholar] [CrossRef]
  24. Zhou, C.; Yamaguchi, M.; Kusaka, G.; Schonholz, C.; Nanda, A.; Zhang, J.H. Caspase Inhibitors Prevent Endothelial Apoptosis and Cerebral Vasospasm in Dog Model of Experimental Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2004, 24, 419–431. [Google Scholar] [CrossRef]
  25. Kwon, M.S.; Woo, S.K.; Kurland, D.B.; Yoon, S.H.; Palmer, A.F.; Banerjee, U.; Iqbal, S.; Ivanova, S.; Gerzanich, V.; Simard, J.M. Methemoglobin Is an Endogenous Toll-Like Receptor 4 Ligand—Relevance to Subarachnoid Hemorrhage. Int. J. Mol. Sci. 2015, 16, 5028–5046. [Google Scholar] [CrossRef]
  26. You, W.-C.; Wang, C.-X.; Pan, Y.-X.; Zhang, X.; Zhou, X.-M.; Zhang, X.-S.; Shi, J.-X.; Zhou, M.-L. Activation of Nuclear Factor-κB in the Brain after Experimental Subarachnoid Hemorrhage and Its Potential Role in Delayed Brain Injury. PLoS ONE 2013, 8, e60290. [Google Scholar] [CrossRef]
  27. Friedrich, V.; Bederson, J.B.; Sehba, F.A. Gender Influences the Initial Impact of Subarachnoid Hemorrhage: An Experimental Investigation. PLoS ONE 2013, 8, e80101. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, T.; Zhang, J.H.; Qin, X. Non-aneurysm subarachnoid hemorrhage in young adults. Acta. Neurochir. Suppl. 2011, 110, 209–213. [Google Scholar] [CrossRef]
  29. Satoh, M.; Parent, A.D.; Zhang, J.H. Inhibitory effect with antisense mitogen-activated protein kinase oligodeoxynucleotide against cerebral vasospasm in rats. Stroke 2002, 33, 775–781. [Google Scholar] [CrossRef] [PubMed]
  30. Zhou, C.; Yamaguchi, M.; Colohan, A.R.; Zhang, J.H. Role of p53 and Apoptosis in Cerebral Vasospasm after Experimental Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2005, 25, 572–582. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, T.; Su, J.; Guo, B.; Wang, K.; Li, X.; Liang, G. Apigenin protects blood–brain barrier and ameliorates early brain injury by inhibiting TLR4-mediated inflammatory pathway in subarachnoid hemorrhage rats. Int. Immunopharmacol. 2015, 28, 79–87. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, G.; Wu, J.; Sun, C.; Qi, M.; Hang, C.; Gong, Y.; Han, X.; Shi, J. Potential role of JAK2 in cerebral vasospasm after experimental subarachnoid hemorrhage. Brain Res. 2008, 1214, 136–144. [Google Scholar] [CrossRef]
  33. Beg, S.S.; Hansen-Schwartz, J.A.; Vikman, P.J.; Xu, C.-B.; Edvinsson, L.I. Protein Kinase C Inhibition Prevents Upregulation of Vascular ETB and 5-HT1B Receptors and Reverses Cerebral Blood Flow Reduction after Subarachnoid Haemorrhage in Rats. J. Cereb. Blood Flow Metab. 2007, 27, 21–32. [Google Scholar] [CrossRef]
  34. Spallone, A. Cerebral vasospasm as a complication of aneurysmal subarachnoid hemorrhage: A brief review. Ital. J. Neurol. Sci. 1985, 6, 19–26. [Google Scholar] [CrossRef]
  35. Chen, S.; Feng, H.; Sherchan, P.; Klebe, D.; Zhao, G.; Sun, X.; Zhang, J.; Tang, J.; Zhang, J.H. Controversies and evolving new mechanisms in subarachnoid hemorrhage. Prog. Neurobiol. 2014, 115, 64–91. [Google Scholar] [CrossRef]
  36. Schebesch, K.-M.; Brawanski, A.; Bele, S.; Schödel, P.; Herbst, A.; Bründl, E.; Kagerbauer, S.M.; Martin, J.; Lohmeier, A.; Stoerr, E.-M.; et al. Neuropeptide Y—An early biomarker for cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Neurol. Res. 2013, 35, 1038–1043. [Google Scholar] [CrossRef]
  37. Wang, Z.; Wang, Y.; Tian, X.; Shen, H.; Dou, Y.; Li, H.; Chen, G. Transient receptor potential channel 1/4 reduces subarachnoid hemorrhage-induced early brain injury in rats via calcineurin-mediated NMDAR and NFAT dephosphorylation. Sci. Rep. 2016, 6, srep33577. [Google Scholar] [CrossRef]
  38. TTokiyoshi, K.; Ohnishi, T.; Nii, Y. Efficacy and toxicity of thromboxane synthetase inhibitor for cerebral vasospasm after subarachnoid hemorrhage. Surg. Neurol. 1991, 36, 112–118. [Google Scholar] [CrossRef]
  39. Beg, S.A.; Hansen-Schwartz, J.A.; Vikman, P.J.; Xu, C.-B.; Edvinsson, L.I. ERK1/2 Inhibition Attenuates Cerebral Blood Flow Reduction and Abolishes ETB and 5-HT1B Receptor Upregulation after Subarachnoid Hemorrhage in Rat. J. Cereb. Blood Flow Metab. 2006, 26, 846–856. [Google Scholar] [CrossRef] [PubMed]
  40. Sugawara, T.; Ayer, R.; Jadhav, V.; Chen, W.; Tsubokawa, T.; Zhang, J.H. Simvastatin attenuation of cerebral vasospasm after subarachnoid hemorrhage in rats via increased phosphorylation of Akt and endothelial nitric oxide synthase. J. Neurosci. Res. 2008, 86, 3635–3643. [Google Scholar] [CrossRef] [PubMed]
  41. Munakata, A.; Naraoka, M.; Katagai, T.; Shimamura, N.; Ohkuma, H. Role of Cyclooxygenase-2 in Relation to Nitric Oxide and Endothelin-1 on Pathogenesis of Cerebral Vasospasm After Subarachnoid Hemorrhage in Rabbit. Transl. Stroke Res. 2016, 7, 220–227. [Google Scholar] [CrossRef] [PubMed]
  42. Roman, R.J.; Renic, M.; Dunn, K.M.J.; Takeuchi, K.; Hacein-Bey, L. Evidence that 20-HETE contributes to the development of acute and delayed cerebral vasospasm. Neurol. Res. 2006, 28, 738–749. [Google Scholar] [CrossRef] [PubMed]
  43. Neuschmelting, V.; Marbacher, S.; Fathi, A.-R.; Jakob, S.M.; Fandino, J. Elevated level of endothelin-1 in cerebrospinal fluid and lack of nitric oxide in basilar arterial plasma associated with cerebral vasospasm after subarachnoid haemorrhage in rabbits. Acta Neurochir. 2009, 151, 795–802. [Google Scholar] [CrossRef]
  44. Takeuchi, K.; Renic, M.; Bohman, Q.C.; Harder, D.R.; Miyata, N.; Roman, R.J. Reversal of delayed vasospasm by an inhibitor of the synthesis of 20-HETE. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H2203–H2211. [Google Scholar] [CrossRef]
  45. Leo, F.; Suvorava, T.; Heuser, S.K.; Li, J.; LoBue, A.; Barbarino, F.; Piragine, E.; Schneckmann, R.; Hutzler, B.; Good, M.E.; et al. Red Blood Cell and Endothelial eNOS Independently Regulate Circulating Nitric Oxide Metabolites and Blood Pressure. Circulation 2021, 144, 870–889. [Google Scholar] [CrossRef]
  46. Cseplo, P.; Vamos, Z.; Ivic, I.; Torok, O.; Toth, A.; Koller, A. The Beta-1-Receptor Blocker Nebivolol Elicits Dilation of Cerebral Arteries by Reducing Smooth Muscle [Ca2+]i. PLoS ONE 2016, 11, e0164010. [Google Scholar] [CrossRef]
  47. Petzold, G.C.; Haack, S.; Halbach, O.v.B.U.; Priller, J.; Lehmann, T.-N.; Heinemann, U.; Dirnagl, U.; Dreier, J.P. Nitric Oxide Modulates Spreading Depolarization Threshold in the Human and Rodent Cortex. Stroke 2008, 39, 1292–1299. [Google Scholar] [CrossRef]
  48. Chen, L.-C.; Hsu, C.; Chiueh, C.C.; Lee, W.-S. Ferrous Citrate Up-Regulates the NOS2 through Nuclear Translocation of NFκB Induced by Free Radicals Generation in Mouse Cerebral Endothelial Cells. PLoS ONE 2012, 7, e46239. [Google Scholar] [CrossRef] [PubMed]
  49. Chang, J.Y.H.; Stamer, W.D.; Bertrand, J.; Read, A.T.; Marando, C.M.; Ethier, C.R.; Overby, D.R. Role of nitric oxide in murine conventional outflow physiology. Am. J. Physiol. Physiol. 2015, 309, C205–C214. [Google Scholar] [CrossRef]
  50. McLeod, D.S.; Hasegawa, T.; Baba, T.; Grebe, R.; D’AUriac, I.G.; Merges, C.; Edwards, M.; Lutty, G.A. From Blood Islands to Blood Vessels: Morphologic Observations and Expression of Key Molecules during Hyaloid Vascular System Development. Investig. Opthalmology Vis. Sci. 2012, 53, 7912–7927. [Google Scholar] [CrossRef]
  51. Yemisci, M.; Gursoy-Ozdemir, Y.; Vural, A.; Can, A.; Topalkara, K.; Dalkara, T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat. Med. 2009, 15, 1031–1037. [Google Scholar] [CrossRef] [PubMed]
  52. Starke, R.M.; Chalouhi, N.; Ali, M.S.; Jabbour, P.M.; Tjoumakaris, S.I.; Gonzalez, L.F.; Rosenwasser, R.H.; Koch, W.J.; Dumont, A.S. The Role of Oxidative Stress in Cerebral Aneurysm Formation and Rupture. Curr. Neurovascular Res. 2013, 10, 247–255. [Google Scholar] [CrossRef]
  53. Förstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837. [Google Scholar] [CrossRef]
  54. Liy, P.M.; Puzi, N.N.A.; Jose, S.; Vidyadaran, S. Nitric oxide modulation in neuroinflammation and the role of mesenchymal stem cells. Exp. Biol. Med. 2021, 246, 2399–2406. [Google Scholar] [CrossRef]
  55. Sonar, S.A.; Lal, G. The iNOS Activity During an Immune Response Controls the CNS Pathology in Experimental Autoimmune Encephalomyelitis. Front. Immunol. 2019, 10, 710. [Google Scholar] [CrossRef] [PubMed]
  56. Zheng, R.; Qin, L.; Li, S.; Xu, K.; Geng, H. CT perfusion-derived mean transit time of cortical brain has a negative correlation with the plasma level of Nitric Oxide after subarachnoid hemorrhage. Acta Neurochir. 2014, 156, 527–533. [Google Scholar] [CrossRef]
  57. Lilla, N.; Hartmann, J.; Koehler, S.; Ernestus, R.-I.; Westermaier, T. Early NO-donor treatment improves acute perfusion deficit and brain damage after experimental subarachnoid hemorrhage in rats. J. Neurol. Sci. 2016, 370, 312–319. [Google Scholar] [CrossRef] [PubMed]
  58. Luettich, A.; Franko, E.; Spronk, D.B.; Lamb, C.; Corkill, R.; Patel, J.; Ezra, M.; Pattinson, K.T.S. Beneficial Effect of Sodium Nitrite on EEG Ischaemic Markers in Patients with Subarachnoid Haemorrhage. Transl. Stroke Res. 2022, 13, 265–275. [Google Scholar] [CrossRef]
  59. Ezra, M.; Garry, P.; Rowland, M.J.; Mitsis, G.D.; Pattinson, K.T. Phase dynamics of cerebral blood flow in subarachnoid haemorrhage in response to sodium nitrite infusion. Nitric Oxide Biol. Chem. 2021, 106, 55–65. [Google Scholar] [CrossRef]
  60. Sehba, F.A.; Schwartz, A.Y.; Chereshnev, I.; Bederson, J.B. Acute Decrease in Cerebral Nitric Oxide Levels after Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2000, 20, 604–611. [Google Scholar] [CrossRef]
  61. Suzuki, Y.; Osuka, K.; Noda, A.; Tanazawa, T.; Takayasu, M.; Shibuya, M.; Yoshida, J. Nitric Oxide Metabolites in the Cisternal Cerebral Spinal Fluid of Patients with Subarachnoid Hemorrhage. Neurosurgery 1997, 41, 807–812. [Google Scholar] [CrossRef]
  62. Springborg, J.B.; Ma, X.; Rochat, P.; Knudsen, G.M.; Amtorp, O.; Paulson, O.B.; Juhler, M.; Olsen, N.V. A single subcutaneous bolus of erythropoietin normalizes cerebral blood flow autoregulation after subarachnoid haemorrhage in rats. Br. J. Pharmacol. 2002, 135, 823–829. [Google Scholar] [CrossRef]
  63. Koide, M.; Bonev, A.D.; Nelson, M.T.; Wellman, G.C. Inversion of neurovascular coupling by subarachnoid blood depends on large-conductance Ca 2+ -activated K + (BK) channels. Proc. Natl. Acad. Sci. USA 2012, 109, E1387–E1395. [Google Scholar] [CrossRef]
  64. Richards, J.; El-Hamamsy, I.; Chen, S.; Sarang, Z.; Sarathchandra, P.; Yacoub, M.H.; Chester, A.H.; Butcher, J.T. Side-Specific Endothelial-Dependent Regulation of Aortic Valve Calcification. Am. J. Pathol. 2013, 182, 1922–1931. [Google Scholar] [CrossRef]
  65. Dreier, J.P.; Major, S.; Manning, A.; Woitzik, J.; Drenckhahn, C.; Steinbrink, J.; Tolias, C.; Oliveira-Ferreira, A.I.; Fabricius, M.; Hartings, J.A.; et al. Cortical spreading ischaemia is a novel process involved in ischaemic damage in patients with aneurysmal subarachnoid haemorrhage. Brain 2009, 132, 1866–1881. [Google Scholar] [CrossRef] [PubMed]
  66. Macdonald, R.L.; Pluta, R.M.; Zhang, J.H. Cerebral vasospasm after subarachnoid hemorrhage: The emerging revolution. Nat. Clin. Pract. Neurol. 2007, 3, 256–263. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, J.; Ye, Z.; Wang, X.; Chang, J.; Yang, M.; Zhong, H.; Hong, F.; Yang, S. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed. Pharmacother. 2018, 97, 423–428. [Google Scholar] [CrossRef]
  68. Saito, A.; Maier, C.M.; Narasimhan, P.; Nishi, T.; Song, Y.S.; Yu, F.; Liu, J.; Lee, Y.-S.; Nito, C.; Kamada, H.; et al. Oxidative Stress and Neuronal Death/Survival Signaling in Cerebral Ischemia. Mol. Neurobiol. 2005, 31, 105–116. [Google Scholar] [CrossRef]
  69. Shin, H.K.; Lee, J.H.; Kim, C.D.; Kim, Y.K.; Hong, J.Y.; Hong, K.W. Prevention of Impairment of Cerebral Blood Flow Autoregulation during Acute Stage of Subarachnoid Hemorrhage by Gene Transfer of Cu/Zn SOD-1 to Cerebral Vessels. J. Cereb. Blood Flow Metab. 2003, 23, 111–120. [Google Scholar] [CrossRef] [PubMed]
  70. Satoh, M.; Date, I.; Nakajima, M.; Takahashi, K.; Iseda, K.; Tamiya, T.; Ohmoto, T.; Ninomiya, Y.; Asari, S. Inhibition of Poly(ADP-Ribose) Polymerase Attenuates Cerebral Vasospasm After Subarachnoid Hemorrhage in Rabbits. Stroke 2001, 32, 225–231. [Google Scholar] [CrossRef]
  71. Leng, L.Z.; Fink, M.E.; Iadecola, C. Spreading Depolarization. Arch. Neurol. 2011, 68, 31–36. [Google Scholar] [CrossRef]
  72. Sanicola, H.W.; Stewart, C.E.; Luther, P.; Yabut, K.; Guthikonda, B.; Jordan, J.D.; Alexander, J.S. Pathophysiology, Management, and Therapeutics in Subarachnoid Hemorrhage and Delayed Cerebral Ischemia: An Overview. Pathophysiology 2023, 30, 420–442. [Google Scholar] [CrossRef] [PubMed]
  73. Mehra, A.; Gomez, F.; Bischof, H.; Diedrich, D.; Laudanski, K. Cortical Spreading Depolarization and Delayed Cerebral Ischemia; Rethinking Secondary Neurological Injury in Subarachnoid Hemorrhage. Int. J. Mol. Sci. 2023, 24, 9883. [Google Scholar] [CrossRef]
  74. Dawson, V.; Dawson, T.; Bartley, D.; Uhl, G.; Snyder, S. Mechanisms of nitric oxide-mediated neurotoxicity in primary brain cultures. J. Neurosci. 1993, 13, 2651–2661. [Google Scholar] [CrossRef]
  75. Belenichev, I.; Popazova, O.; Bukhtiyarova, N.; Savchenko, D.; Oksenych, V.; Kamyshnyi, O. Modulating Nitric Oxide: Implications for Cytotoxicity and Cytoprotection. Antioxidants 2024, 13, 504. [Google Scholar] [CrossRef]
  76. Solodov, A.A.; Petrikov, S.S.; Klychnnikova, E.V.; Tazina, E.V.; Krylov, V.V.; Godkov, M.A.; Khamidova, L.T. Effect of normobaric hyperoxia on cerebral oxygenation, metabolism and oxidative stress in patients with subarachnoid hemorrhage caused by intracranial aneurysm rupture. Anesteziol. Reanimatol. 2013, 4, 66–71. [Google Scholar]
  77. Sasaki, T.; Wakai, S.; Asano, T.; Watanabe, T.; Kirino, T.; Sano, K. The effect of a lipid hydroperoxide of arachidonic acid on the canine basilar artery. An experimental study on cerebral vasospasm. J. Neurosurg. 1981, 54, 357–365. [Google Scholar] [CrossRef]
  78. Lan, S.H.; Lai, W.T.; Zheng, S.Y.; Yang, L.; Fang, L.C.; Zhou, L.; Tang, B.; Duan, J.; Hong, T. Upregulation of Connexin 40 Mediated by Nitric Oxide Attenuates Cerebral Vasospasm After Subarachnoid Hemorrhage via the Nitric Oxide-Cyclic Guanosine Monophosphate-Protein Kinase G Pathway. World Neurosurg. 2020, 136, e476–e486. [Google Scholar] [CrossRef]
  79. Bosche, B.; Graf, R.; Ernestus, R.; Dohmen, C.; Reithmeier, T.; Brinker, G.; Strong, A.J.; Dreier, J.P.; Woitzik, J. Recurrent spreading depolarizations after subarachnoid hemorrhage decreases oxygen availability in human cerebral cortex. Ann. Neurol. 2010, 67, 607–617. [Google Scholar] [CrossRef] [PubMed]
  80. Keyrouz, S.G.; Diringer, M.N. Clinical review: Prevention and therapy of vasospasm in subarachnoid hemorrhage. Crit. Care 2007, 11, 220. [Google Scholar] [CrossRef] [PubMed]
  81. Starke, R.M.; Kim, G.H.; Komotar, R.J.; Hickman, Z.L.; Black, E.M.; Rosales, M.B.; Kellner, C.P.; Hahn, D.K.; Otten, M.L.; Edwards, J. Endothelial Nitric Oxide Synthase Gene Single Nucleotide Polymorphism Predicts Cerebral Vasospasm following Aneurysmal Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2008, 28, 1204–1211. [Google Scholar] [CrossRef]
  82. Faraco, G.; Moraga, A.; Moore, J.; Anrather, J.; Pickel, V.M.; Iadecola, C. Circulating Endothelin-1 Alters Critical Mechanisms Regulating Cerebral Microcirculation. Hypertension 2013, 62, 759–766. [Google Scholar] [CrossRef] [PubMed]
  83. Sabri, M.; Ai, J.; Knight, B.; Tariq, A.; Jeon, H.; Shang, X.; Marsden, P.A.; Macdonald, R.L. Uncoupling of Endothelial Nitric Oxide Synthase after Experimental Subarachnoid Hemorrhage. J. Cereb. Blood Flow Metab. 2011, 31, 190–199. [Google Scholar] [CrossRef]
  84. Li, S.; Tian, Y.; Huang, X.; Zhang, Y.; Wang, D.; Wei, H.; Dong, J.; Jiang, R.; Zhang, J. Intravenous transfusion of endothelial colony-forming cells attenuates vascular degeneration after cerebral aneurysm induction. Brain Res. 2014, 1593, 65–75. [Google Scholar] [CrossRef]
  85. Chang, C.-Z.; Wu, S.-C.; Chang, C.-M.; Lin, C.-L.; Kwan, A.-L. Arctigenin, a Potent Ingredient of Arctium lappa L., Induces Endothelial Nitric Oxide Synthase and Attenuates Subarachnoid Hemorrhage-Induced Vasospasm through PI3K/Akt Pathway in a Rat Model. BioMed Res. Int. 2015, 2015, 490209. [Google Scholar] [CrossRef]
  86. Cho, H.G.; Shin, H.K.; Shin, Y.W.; Lee, J.H.; Hong, K.W. Role of nitric oxide in the CBF autoregulation during acute stage after subarachnoid haemorrhage in rat pial artery. Fundam. Clin. Pharmacol. 2003, 17, 563–573. [Google Scholar] [CrossRef]
  87. Maddahi, A.; Ansar, S.; Chen, Q.; Edvinsson, L. Blockade of the MEK/ERK Pathway with a Raf Inhibitor Prevents Activation of Pro-Inflammatory Mediators in Cerebral Arteries and Reduction in Cerebral Blood Flow after Subarachnoid Hemorrhage in a Rat Model. J. Cereb. Blood Flow Metab. 2011, 31, 144–154. [Google Scholar] [CrossRef] [PubMed]
  88. Vikman, P.; Ansar, S.; Henriksson, M.; Stenman, E.; Edvinsson, L. Cerebral ischemia induces transcription of inflammatory and extracellular-matrix-related genes in rat cerebral arteries. Exp. Brain Res. 2007, 183, 499–510. [Google Scholar] [CrossRef]
  89. Griessenauer, C.J.; Starke, R.M.; Foreman, P.M.; Hendrix, P.; Harrigan, M.R.; Fisher, W.S.; Vyas, N.A.; Lipsky, R.H.; Lin, M.; Walters, B.C.; et al. Associations between endothelin polymorphisms and aneurysmal subarachnoid hemorrhage, clinical vasospasm, delayed cerebral ischemia, and functional outcome. J. Neurosurg. 2018, 128, 1311–1317. [Google Scholar] [CrossRef]
  90. Yeung, P.K.; Shen, J.; Chung, S.S.; Chung, S.K. Targeted over-expression of endothelin-1 in astrocytes leads to more severe brain damage and vasospasm after subarachnoid hemorrhage. BMC Neurosci. 2013, 14, 131. [Google Scholar] [CrossRef] [PubMed]
  91. Macdonald, R.L.; Kassell, N.F.; Mayer, S.; Ruefenacht, D.; Schmiedek, P.; Weidauer, S.; Frey, A.; Roux, S.; Pasqualin, A. CONSCIOUS-1 Investigators Clazosentan to Overcome Neurological Ischemia and Infarction Occurring After Subarachnoid Hemorrhage (CONSCIOUS-1): Randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke 2008, 39, 3015–3021. [Google Scholar] [CrossRef]
  92. Galiè, N.; Olschewski, H.; Oudiz, R.J.; Torres, F.; Frost, A.; Ghofrani, H.A.; Badesch, D.B.; McGoon, M.D.; McLaughlin, V.V.; Roecker, E.B.; et al. Ambrisentan for the Treatment of Pulmonary Arterial Hypertension. Circulation 2008, 117, 3010–3019. [Google Scholar] [CrossRef]
  93. Kim, M.; Jeon, H.; Chung, Y.; Lee, S.U.; Park, W.; Park, J.C.; Ahn, J.S.; Lee, S. Efficacy of Acetylcysteine and Selenium in Aneurysmal Subarachnoid Hemorrhage Patients: A Prospective, Multicenter, Single Blind Randomized Controlled Trial. J. Korean Med. Sci. 2023, 38, e161. [Google Scholar] [CrossRef]
  94. Tsai, I.J.; Croft, K.D.; Puddey, I.B.; Beilin, L.J.; Barden, A. 20-Hydroxyeicosatetraenoic acid synthesis is increased in human neutrophils and platelets by angiotensin II and endothelin-1. Am. J. Physiol. Circ. Physiol. 2011, 300, H1194–H1200. [Google Scholar] [CrossRef]
  95. Miyata, N.; Seki, T.; Tanaka, Y.; Omura, T.; Taniguchi, K.; Doi, M.; Bandou, K.; Kametani, S.; Sato, M.; Okuyama, S.; et al. Beneficial Effects of a New 20-Hydroxyeicosatetraenoic Acid Synthesis Inhibitor, TS-011 [N-(3-Chloro-4-morpholin-4-yl) Phenyl-N′-hydroxyimido Formamide], on Hemorrhagic and Ischemic Stroke. J. Pharmacol. Exp. Ther. 2005, 314, 77–85. [Google Scholar] [CrossRef]
  96. Vergouwen, M.D.; de Haan, R.J.; Vermeulen, M.; Roos, Y.B. Statin Treatment and the Occurrence of Hemorrhagic Stroke in Patients With a History of Cerebrovascular Disease. Stroke 2008, 39, 497–502. [Google Scholar] [CrossRef]
  97. Vergouwen, M.D.; de Haan, R.J.; Vermeulen, M.; Roos, Y.B. Effect of Statin Treatment on Vasospasm, Delayed Cerebral Ischemia, and Functional Outcome in Patients With Aneurysmal Subarachnoid Hemorrhage. Stroke 2010, 41, e47–e52. [Google Scholar] [CrossRef] [PubMed]
  98. Khurana, V.G.; Smith, L.A.; Baker, T.A.; Eguchi, D.; O’brien, T.; Katusic, Z.S. Protective Vasomotor Effects of In Vivo Recombinant Endothelial Nitric Oxide Synthase Gene Expression in a Canine Model of Cerebral Vasospasm. Stroke 2002, 33, 782–789. [Google Scholar] [CrossRef]
  99. Zhao, Y.D.; Courtman, D.W.; Deng, Y.; Kugathasan, L.; Zhang, Q.; Stewart, D.J. Rescue of Monocrotaline-Induced Pulmonary Arterial Hypertension Using Bone Marrow–Derived Endothelial-Like Progenitor Cells: Efficacy of combined cell and eNOS gene therapy in established disease. Circ. Res. 2005, 96, 442–450. [Google Scholar] [CrossRef]
  100. Taneja, G.; Sud, A.; Pendse, N.; Panigrahi, B.; Kumar, A.; Sharma, A.K. Nano-medicine and Vascular Endothelial Dysfunction: Options and Delivery Strategies. Cardiovasc. Toxicol. 2019, 19, 1–12. [Google Scholar] [CrossRef]
  101. Dutta, P.; Hoyer, F.F.; Grigoryeva, L.S.; Sager, H.B.; Leuschner, F.; Courties, G.; Borodovsky, A.; Novobrantseva, T.; Ruda, V.M.; Fitzgerald, K.; et al. Macrophages retain hematopoietic stem cells in the spleen via VCAM-1. J. Exp. Med. 2015, 212, 497–512. [Google Scholar] [CrossRef]
  102. Stoodley, M.; Weihl, C.C.; Zhang, Z.-D.; Lin, G.; Johns, L.M.; Kowalczuk, A.; Ghadge, G.; Roos, R.P.; Macdonald, R.L. Effect of Adenovirus-mediated Nitric Oxide Synthase Gene Transfer on Vasospasm after Experimental Subarachnoid Hemorrhage. Neurosurgery 2000, 46, 1193–1203. [Google Scholar] [CrossRef] [PubMed]
  103. Onoue, H.; Tsutsui, M.; Smith, L.; Stelter, A.; O’bRien, T.; Katusic, Z.S. Expression and Function of Recombinant Endothelial Nitric Oxide Synthase Gene in Canine Basilar Artery After Experimental Subarachnoid Hemorrhage. Stroke 1998, 29, 1959–1966. [Google Scholar] [CrossRef] [PubMed]
  104. Medana, C.; Di Stilo, A.; Visentin, S.; Fruttero, R.; Gasco, A.; Ghigo, D.; Bosia, A. NO donor and biological properties of different benzofuroxans. Pharm Res. 1999, 16, 956–960. [Google Scholar] [CrossRef]
  105. Bussygina, O.G.; Pyatakova, N.V.; Khropov, Y.V.; Ovchinnikov, I.V.; Makhova, N.N.; Severina, I.S. Benzodifuroxan as an NO-dependent activator of soluble guanylate cyclase and a novel highly effective inhibitor of platelet aggregation. Biochemistry 2000, 65, 457–462. [Google Scholar] [PubMed]
  106. Macdonald, R.L. Clazosentan: An endothelin receptor antagonist for treatment of vasospasm after subarachnoid hemorrhage. Expert Opin. Investig. Drugs 2008, 17, 1761–1767. [Google Scholar] [CrossRef]
  107. Iglarz, M.; Binkert, C.; Morrison, K.; Fischli, W.; Gatfield, J.; Treiber, A.; Weller, T.; Bolli, M.H.; Boss, C.; Buchmann, S.; et al. Pharmacology of Macitentan, an Orally Active Tissue-Targeting Dual Endothelin Receptor Antagonist. J. Pharmacol. Exp. Ther. 2008, 327, 736–745. [Google Scholar] [CrossRef]
  108. Heerspink, H.J.L.; Radhakrishnan, J.; Alpers, C.E.; Barratt, J.; Bieler, S.; Diva, U.; Inrig, J.; Komers, R.; Mercer, A.; Noronha, I.L.; et al. Sparsentan in patients with IgA nephropathy: A prespecified interim analysis from a randomised, double-blind, active-controlled clinical trial. Lancet 2023, 401, 1584–1594. [Google Scholar] [CrossRef]
  109. Ehlers, J.P. PER-001, a long-acting endothelin antagonist intravitreal implant, improved structure and visual function in patients with diabetic retinopathy. In Proceedings of the 43rd Annual Meeting of the American Society of Retina Specialists, Long Beach, CA, USA, 30 July–2 August 2025. [Google Scholar]
  110. Froogh, G.; Garcia, V.; Laniado Schwartzman, M. The CYP/20-HETE/GPR75 axis in hypertension. Adv. Pharmacol. 2022, 94, 1–25. [Google Scholar] [CrossRef]
  111. Elmarakby, A.A.; Morsing, P.; Pollock, D.M. Enalapril attenuates endothelin-1-induced hypertension via increased kinin survival. Am. J. Physiol. Heart Circ. Physiol. 2003, 284, 1899–1903. [Google Scholar] [CrossRef]
  112. Meis, T.; Behr, J. Riociguat for the treatment of pulmonary hypertension. Expert Opin. Pharmacother. 2014, 15, 2419–2427. [Google Scholar] [CrossRef]
  113. Kehl, F.; Cambj-Sapunar, L.; Maier, K.G.; Miyata, N.; Kametani, S.; Okamoto, H.; Hudetz, A.G.; Schulte, M.L.; Zagorac, D.; Harder, D.R.; et al. 20-HETE contributes to the acute fall in cerebral blood flow after subarachnoid hemorrhage in the rat. Am. J. Physiol. Circ. Physiol. 2002, 282, H1556–H1565. [Google Scholar] [CrossRef]
  114. Cambj-Sapunar, L.; Yu, M.; Harder, D.R.; Roman, R.J. Contribution of 5-Hydroxytryptamine 1B Receptors and 20-Hydroxyeiscosatetraenoic Acid to Fall in Cerebral Blood Flow After Subarachnoid Hemorrhage. Stroke 2003, 34, 1269–1275. [Google Scholar] [CrossRef]
  115. Benter, I.; Yousif, M.; Canatan, H.; Akhtar, S. Inhibition of Ca/calmodulin-dependent protein kinase II, RAS-GTPase and 20-hydroxyeicosatetraenoic acid attenuates the development of diabetes-induced vascular dysfunction in the rat carotid artery. Pharmacol. Res. 2005, 52, 252–257. [Google Scholar] [CrossRef]
  116. Wang, C.; Zhao, X.; Mao, S.; Wang, Y.; Cui, X.; Pu, Y. Management of SAH with traditional Chinese medicine in China. Neurol. Res. 2006, 28, 436–444. [Google Scholar] [CrossRef]
  117. Seo, Y.; Lee, H.-G.; Jin, C.; Yang, S.-B.; Cho, S.-Y.; Park, S.-U.; Jung, W.-S.; Moon, S.-K.; Park, J.-M.; Ko, C.-N.; et al. Herbal medicines for the prevention and treatment of cerebral vasospasm after subarachnoid hemorrhage: A protocol for systematic review and meta-analysis. Medicine 2020, 99, e23388. [Google Scholar] [CrossRef]
  118. Mouratoglou, S.A.; Arvanitaki, A.; Papadopoulos, G.; Souza, R.; Giannakoulas, G. Pulmonary arterial hypertension treatment. A new era. Int. J. Cardiol. Congenit. Heart Dis. 2025, 21, 100594. [Google Scholar] [CrossRef]
  119. Macdonald, R.L. Delayed neurological deterioration after subarachnoid haemorrhage. Nat. Rev. Neurol. 2013, 10, 44–58. [Google Scholar] [CrossRef]
  120. Cho, S.S.; Kim, S.-E.; Kim, H.C.; Kim, W.J.; Jeon, J.P. Clazosentan for Aneurysmal Subarachnoid Hemorrhage: An Updated Meta-Analysis with Trial Sequential Analysis. World Neurosurg. 2019, 123, 418–424.e3. [Google Scholar] [CrossRef]
  121. Ezra, M.; Franko, E.; Spronk, D.B.; Lamb, C.; Okell, T.W.; Pattinson, K.T. Trial of the cerebral perfusion response to sodium nitrite infusion in patients with acute subarachnoid haemorrhage using arterial spin labelling MRI. Nitric Oxide Biol. Chem. 2024, 153, 50–60. [Google Scholar] [CrossRef]
  122. Achrol, A.S.; Steinberg, G.K. Personalized Medicine in Cerebrovascular Neurosurgery: Precision Neurosurgical Management of Cerebral Aneurysms and Subarachnoid Hemorrhage. Front. Surg. 2016, 3, 34. [Google Scholar] [CrossRef]
  123. Durgin, B.G.; Hahn, S.A.; Schmidt, H.M.; Miller, M.P.; Hafeez, N.; Mathar, I.; Freitag, D.; Sandner, P.; Straub, A.C. Loss of smooth muscle CYB5R3 amplifies angiotensin II–induced hypertension by increasing sGC heme oxidation. J. Clin. Investig. 2019, 4, e129183. [Google Scholar] [CrossRef]
  124. Moustakas, D.; Mani, I.; Pouliakis, A.; Iacovidou, N.; Xanthos, T. The Effects of IRL-1620 in Post-ischemic Brain Injury: A Systematic Review and Meta-analysis of Experimental Studies. Neurocritical Care 2024, 41, 665–680. [Google Scholar] [CrossRef]
  125. Abu-Soud, H.M.; Presta, A.; Mayer, B.; Stuehr, D.J. Analysis of Neuronal NO Synthase under Single-Turnover Conditions: Conversion of Nω-Hydroxyarginine to Nitric Oxide and Citrulline. Biochemistry 1997, 36, 10811–10816. [Google Scholar] [CrossRef]
  126. Kapil, V.; Khambata, R.S.; Jones, D.A.; Rathod, K.; Primus, C.; Massimo, G.; Fukuto, J.M.; Ahluwalia, A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol. Rev. 2020, 72, 692–766. [Google Scholar] [CrossRef]
  127. van Faassen, E.E.; Bahrami, S.; Feelisch, M.; Hogg, N.; Kelm, M.; Kim-Shapiro, D.B.; Kozlov, A.V.; Li, H.; Lundberg, J.O.; Mason, R.; et al. Nitrite as regulator of hypoxic signaling in mammalian physiology. Med. Res. Rev. 2009, 29, 683–741. [Google Scholar] [CrossRef]
  128. Timilsina, A.; Dong, W.; Hasanuzzaman, M.; Liu, B.; Hu, C. Nitrate–Nitrite–Nitric Oxide Pathway: A Mechanism of Hypoxia and Anoxia Tolerance in Plants. Int. J. Mol. Sci. 2022, 23, 11522. [Google Scholar] [CrossRef]
  129. Fathi, A.R.; Pluta, R.M.; Bakhtian, K.D.; Qi, M.; Lonser, R.R. Reversal of cerebral vasospasm via intravenous sodium nitrite after subarachnoid hemorrhage in primates: Laboratory investigation. J. Neurosurg. 2011, 115, 1213–1220. [Google Scholar] [CrossRef]
  130. Oldfield, E.H.; Loomba, J.J.; Monteith, S.J.; Crowley, R.W.; Medel, R.; Gress, D.R.; Kassell, N.F.; Dumont, A.S.; Sherman, C. Safety and pharmacokinetics of sodium nitrite in patients with subarachnoid hemorrhage: A Phase IIA study. J. Neurosurg. 2013, 119, 634–641. [Google Scholar] [CrossRef]
  131. Larsen, F.J.; Schiffer, T.A.; Borniquel, S.; Sahlin, K.; Ekblom, B.; Lundberg, J.O.; Weitzberg, E. Dietary Inorganic Nitrate Improves Mitochondrial Efficiency in Humans. Cell Metab. 2011, 13, 149–159. [Google Scholar] [CrossRef]
Figure 1. Depiction of the relevant NO/ET-1 axis pathophysiological pathways for the development of DCI and DIND following SAH. (A) Molecular pathogenesis initiated by SAH. The pathophysiological cascade is characterized by synergistic interactions: the initial insult involves massive scavenging of NO and a surge in reactive oxygen species (ROS), which directly damage endothelium and induce eNOS uncoupling. These processes induce the upregulation of 20-HETE, a key mediator that suppresses residual cGMP-dependent vasodilation. In parallel, ROS and other inflammatory mediators co-activate MAPK and PI3K/Akt pathways, which further exacerbate the imbalance by enhancing vasoconstrictor gene expression (e.g., ET-1) and altering NOS activity, thereby cementing the vicious cycle. (B) Pathophysiological cascade in the microvasculature. (C) Representative pharmacotherapies for axis recalibration, including hypoxia-activated nitric oxide donors, ET-1 receptor antagonists (e.g., clazosentan), and multi-pathway agents such as 20-HETE synthesis inhibitors (e.g., HET0016) that target convergent mechanisms of vasospasm. Figure was prepared using Servier Medical Art (https://smart.servier.com/ (accessed on 4 September 2025)), licensed under CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/ (accessed on 4 September 2025)).
Figure 1. Depiction of the relevant NO/ET-1 axis pathophysiological pathways for the development of DCI and DIND following SAH. (A) Molecular pathogenesis initiated by SAH. The pathophysiological cascade is characterized by synergistic interactions: the initial insult involves massive scavenging of NO and a surge in reactive oxygen species (ROS), which directly damage endothelium and induce eNOS uncoupling. These processes induce the upregulation of 20-HETE, a key mediator that suppresses residual cGMP-dependent vasodilation. In parallel, ROS and other inflammatory mediators co-activate MAPK and PI3K/Akt pathways, which further exacerbate the imbalance by enhancing vasoconstrictor gene expression (e.g., ET-1) and altering NOS activity, thereby cementing the vicious cycle. (B) Pathophysiological cascade in the microvasculature. (C) Representative pharmacotherapies for axis recalibration, including hypoxia-activated nitric oxide donors, ET-1 receptor antagonists (e.g., clazosentan), and multi-pathway agents such as 20-HETE synthesis inhibitors (e.g., HET0016) that target convergent mechanisms of vasospasm. Figure was prepared using Servier Medical Art (https://smart.servier.com/ (accessed on 4 September 2025)), licensed under CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/ (accessed on 4 September 2025)).
Futurepharmacol 05 00059 g001aFuturepharmacol 05 00059 g001b
Table 1. Pharmacological interventions targeting the NO/ET-1 axis.
Table 1. Pharmacological interventions targeting the NO/ET-1 axis.
StrategyAgentsMechanismRef.
NO DonorsSodium nitrite, L-ArginineHypoxia-triggered NO release; sGC activationØstergaard L et al., 2013 [14]; Lilla N et al., 2016 [57]
ET-1 AntagonistsClazosentan, BosentanSelective ETA blockade → ROS/eNOS uncoupling reversalMacdonald RL et al., 2008 [91];
Galiè N et al., 2008 [92]
NO Bioavailability EnhancersNAC, GlutathioneScavenge ROS → reduce NO degradation, potentially reversing eNOS uncouplingKim M et al., 2023 [93]
Multi-Target AgentsHET0016, TS-011Multi-component synergy → restore NO-cGMP signaling, NO synthesis↑Tsai IJ et al., 2011 [94]; Miyata N et al., 2005 [95]
Adjuvant TherapiesStatins, ErythropoietinAkt-eNOS phosphorylation → NO synthesis ↑Vergouwen MD et al., 2008 [96]; Vergouwen MD et al., 2010 [97]
Emerging ApproachesNOS gene therapy, NanocarriersTargeted eNOS delivery/activationKhurana VG et al., 2002 [98]; Zhao YD et al., 2005 [99]; Taneja G et al., 2019 [100]
Table 2. Representative multi-target agents modulating NO/ET-1 homeostasis.
Table 2. Representative multi-target agents modulating NO/ET-1 homeostasis.
Drug ClassRepresentativesMechanismRef.
ACEi/ARBsEnalaprilInhibit Ang II → ↓ET-1 synthesis + ↑NO releaseElmarakby AA et al., 2003 [111]
sGC StimulatorsRiociguatDirectly activate sGC (NO-independent) → ↑cGMPMeis T et al., 2014 [112]
20-HETE InhibitorsHET0016, TS-011Inhibit CYP4A → restore NO-cGMP signaling, reverse pericyte constrictionKehl F et al., 2002 [113]; Cambj-Sapunar L et al., 2003 [114]; Tsai IJ et al., 2011 [94]; Benter IF et al., 2005 [115]; Takeuchi K et al., 2005 [44]
Herbal FormulationsLigusticum chuanxiongMulti-component synergy: ↓ET-1/IL-5 + ↑NOWang C et al., 2006 [116]; Seo Y et al., 2020 [117]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Becker, K.; Lu, K. Bidirectional Regulation of Nitric Oxide and Endothelin-1 in Cerebral Vasospasm: Mechanisms and Therapeutic Perspectives. Future Pharmacol. 2025, 5, 59. https://doi.org/10.3390/futurepharmacol5040059

AMA Style

Becker K, Lu K. Bidirectional Regulation of Nitric Oxide and Endothelin-1 in Cerebral Vasospasm: Mechanisms and Therapeutic Perspectives. Future Pharmacology. 2025; 5(4):59. https://doi.org/10.3390/futurepharmacol5040059

Chicago/Turabian Style

Becker, Katrin, and Kaihui Lu. 2025. "Bidirectional Regulation of Nitric Oxide and Endothelin-1 in Cerebral Vasospasm: Mechanisms and Therapeutic Perspectives" Future Pharmacology 5, no. 4: 59. https://doi.org/10.3390/futurepharmacol5040059

APA Style

Becker, K., & Lu, K. (2025). Bidirectional Regulation of Nitric Oxide and Endothelin-1 in Cerebral Vasospasm: Mechanisms and Therapeutic Perspectives. Future Pharmacology, 5(4), 59. https://doi.org/10.3390/futurepharmacol5040059

Article Metrics

Back to TopTop