IMR Press / JIN / Volume 19 / Issue 4 / DOI: 10.31083/j.jin.2020.04.370
Open Access Mini-Review
Can we improve the prediction of complications and outcome in aneurysmal subarachnoid hemorrhage? The clinical implications of serum proteomics
Show Less
1 Department of Neurosurgery, American University of Beirut Medical Center, Beirut, Lebanon
2 Faculty of Medicine, Lebanese University, Beirut, Lebanon
*Correspondence: ts18@aub.edu.lb (Tarek Sunna)
J. Integr. Neurosci. 2020, 19(4), 711–717; https://doi.org/10.31083/j.jin.2020.04.370
Submitted: 3 November 2020 | Revised: 29 November 2020 | Accepted: 14 December 2020 | Published: 30 December 2020
Copyright: © 2020 Bsat et al. Published by IMR Press.
This is an open access article under the CC BY 4.0 license ( https://creativecommons.org/licenses/by/4.0/).
Abstract

Aneurysmal subarachnoid hemorrhage is a devastating condition, often leading to a debilitating outcome. Delayed ischemic neurological deficits are considered the feared sequelae. Proteomics is a large-scale study of proteins incorporating structural and functional properties in complex biological fluids. Analysis of proteomes has led to identifying relevant complex proteins related to specific pathophysiological processes reflecting the severity and extent of diseases. Proteomics has evolved in the past few years; more biomarkers are deemed clinically relevant to diagnose, monitor, and define prognosis in patients with aneurysmal subarachnoid hemorrhage. Despite the absence of candidate biomarkers in the clinical routine, many have shown promising results. The complexity of proteins implicated in aneurysmal subarachnoid hemorrhage rendered these biomarkers’ clinical use paved with various pitfalls and technical difficulties, especially when data about the perfect timing and values are lacking. We review the latest literature concerning serum proteomics and their clinical utility regarding the prediction of cerebral vasospasm and other complications of aneurysmal subarachnoid hemorrhage, as well as the clinical outcome. Future prospective studies will allow changing the disease’s course, label patients according to their prognosis to provide earlier and better management and improve outcomes.

Keywords
Proteome
aneurysm
subarachnoid hemorrhage
vasospasm
delayed cerebral ischemia
1. Introduction

Among the vast etiologies of strokes, aneurysmal subarachnoid hemorrhage (aSAH) accounts for about 2-5% of all stroke cases (Lovelock et al., 2010). The occurrence of multiple post-hemorrhagic complications such as the initial parenchymal damage of the subarachnoid hemorrhage (De Marchis et al., 2015; Eriksen et al., 2019; Hartings et al., 2017; Sarrafzadeh et al., 2010), delayed cerebral ischemia (DCI), hydrocephalus, epilepsy, and systemic infections, constitute the reason for poor outcome after aSAH (Frontera et al., 2008). DCI is the main predictor of clinical outcome in aSAH and was believed to come secondary to cerebral vasospasm (CVS) in 20-30% of cases (Haley et al., 1992). More recently, it is found to be a more complex multifactorial phenomenon. A spreading depolarization precedes the decreased cerebral blood flow and partial tissue pressure of oxygen in the microcirculation, which leads to stroke and subsequent cytotoxic edema (Lückl et al., 2018). This acute disturbance in the microcirculation can be merely due to neuronal and glial depolarization, which release high potent vasoconstrictors (Dreier, 2011). This was deduced from the alternating patterns of hyperperfusion-hypoperfusion during the acute depolarization waves (Lückl et al., 2018; Minhas et al., 2003), which cannot be the mere result of sustained vasoconstriction of large arteries (Minhas et al., 2003). The most used scales to predict prognosis and outcome in patients with aSAH are Hunt and Hess (1968) and the World Federation of Neurological Surgeons (WFNS). Since both are clinical scores and given the dire consequences of aSAH complications, there is a need for an objective test that is non-invasive, simple and reliable, predicting these complications and improving the timely delivery of appropriate treatment.

Serum biology involves genomics/transcriptomics, proteomics, glycomics, lipidomics, metabolomics and degradomics (Jean Beltran et al., 2017). Most recent data has demonstrated the presence of a metabolic crisis correlating with various clinical outcomes, and temporal correlations have been confirmed as early as 15 minutes after subarachnoid hemorrhage (Gewirtz et al., 1999).

The term proteomics describes the study of a complete set of proteins existent in a cell, organ, or organism at a specific time (Wilkins et al., 1996). It’s a large-scale analysis of proteins, found as multiprotein complexes, or all proteins encoded by the genome, constituting the proteome (Pandey and Mann, 2000). Biomarker discovery is a very challenging procedure due to the complexity of the body fluids and the wide variety of protein concentrations found (Seibert, 2005). Significant workup has been done regarding the identification, validation, and clinical correlation of different protein biomarkers to measure the severity of strokes and traumatic brain injury (Mrozek et al., 2014). Protein micro identification and sequencing have evolved substantially using mass spectrometry, a very sensitive analytical method (Shevchenko et al., 1999). Fig. 1 presents the location of serum proteomics involved in aSAH. It also highlights the consequences of disrupting the blood-brain barrier and its implications (Lublinsky et al., 2019). It is important to find proteomes that can be deemed predictors of DCI and other complications of aSAH to treat them timely and correctly. None of these biomarkers have reached routine clinical use due to a lack of enough integrated data about specific timing, techniques, and clinical significance. This article reviews recent studies regarding serum proteomics as mediators of the disease and predictors of complications and outcome, as a bridge towards diagnostic workup and treatment guidance. Results are summarized in Table 1 and Table 2.

Fig. 1.

A schematic illustration showing proteomes related to aSAH, and their location inside neurons, astrocytes, and blood vessel. The disruption of the blood-brain barrier allows the pouring of biomarkers with subsequent inflammation of the surrounding brain parenchyma. ET-1: Endothelin-1. TRX: Thioredoxin. NSE: Neuron-specific enolase. IL-6: Interleukin-6. CRP: C-reactive Protein.

Table 1.Proteomics that are significantly related to clinical vasospasm and DIND, with their serum value and time of sample collection.
𝑩 𝒊 𝒐 𝒎 𝒂 𝒓 𝒌 𝒆 𝒓 𝒔 𝒕 𝒖 𝒅 𝒚 / 𝒚 𝒆 𝒂 𝒓 𝒍 𝒐 𝒘 𝒓 𝒊 𝒔 𝒌 𝒐 𝒇 𝒉 𝒊 𝒈 𝒉 𝒓 𝒊 𝒔 𝒌 𝒐 𝒇 𝒕 𝒊 𝒎 𝒆 𝒐 𝒇
𝒗 𝒂 𝒔 𝒐 𝒔 𝒑 𝒂 𝒔 𝒎 𝒂 𝒏 𝒅 𝑫 𝑰 𝑵 𝑫 𝒗 𝒂 𝒔 𝒐 𝒔 𝒑 𝒂 𝒔 𝒎 𝒂 𝒏 𝒅 𝑫 𝑰 𝑵 𝑫 𝒔 𝒂 𝒎 𝒑 𝒍 𝒆 𝒕 𝒂 𝒌 𝒆 𝒏
Endothelin-1 Juvela (2000)  median 2 pg/mL median 2.7 pg/mL within 24 hours from admission
Nesfatin-1 Wei et al. (2018) Median 8.84 ng/mL Median 11.73 ng/mL On admission
C-reactive protein Romero et al. (2014) Mean 3 mg/L Mean 16 mg/L Between 3 r d and 4 t h day
Myeloperoxidase Lim et al. (2012) Mean 59.4 ng/mL Mean 115.5 ng/mL 2 or more days prior to vasospasm
Interleukin-6 Chaudhry et al. (2017) Mean 1.0 ± 0.2 (log IL-6 pg/mL) Mean 1.5 ± 0.2 (log IL-6 pg/mL) Days 3 7 9 post aSAH
Table 2.Proteomics is significantly related to a bad outcome, with their serum value and time of sample collection.
𝑩 𝒊 𝒐 𝒎 𝒂 𝒓 𝒌 𝒆 𝒓 study/year Good outcome bad outcome time of sample taken
Nesfatin-1 Wei et al. (2018) Median 9.11 ng/mL Median 11.34 ng/mL On admission
Thioredoxin Dai et al. (2016) < 25.1 ng/mL > 25.1 ng/mL On admission
C-reactive protein Romero et al. (2014) Mean 4 mg/L Mean 8-10 mg/L Between 3 r d and 4 t h day
Copeptin Fung et al. (2013) Median 6.8 pmol/L Median 26.3 pmol/L At admission
Albumin Kapoor et al. (2018) > 3.5 mg/dL < 3.5 mg/dL At admission
Neuron-specific enolase Tawk et al. (2016) < 15 ng/mL > 15 ng/mL Within a mean of 25.2 hours
S100B Moritz et al. (2010) Mean < 0.17 µg/L Mean > 0.17 µg/L Sampling during 15 days post aSAH
Interleukin-6 Chaudhry et al. (2017) Mean 1.0 ± 0.2 (log IL-6 pg/mL) Mean 1.5 ± 0.2 (log IL-6 pg/mL) Days 3 7 9 post aSAH
2. Endothelin-1 (ET-1)

The most potent endogenous vasoconstrictor, endothelin-1, is a 21 amino acid peptide produced by endothelial cells (Suhardja, 2004). Most of the studies regarding endothelin-1 levels and its prediction of cerebral vasospasm were done on Cerebrospinal Fluid (CSF) samples. Very few articles mentioned plasma ET-1 as a potential predictor of vasospasm and DCI, but most of them were descriptive or done on animals. A study involved 70 patients with a median age of 45.8 years, ET values were measured in samples obtained within 24 hours either before or after angiography were higher in patients with angiographic evidence of severe vasospasm (2.7 ± 0.4 pg/mL in 13 patients compared with 2 ± 0.2 pg/mL in 34 patients; P = 0.078); the values were also higher in patients with angiographic evidence of diffuse moderate-to-severe vasospasm (2.6 ± 0.3 pg/mL) than in others (Juvela, 2000). Since plasma ET-1 shows promising results, more studies should be done on plasma levels since it will be more feasible, especially in critically ill patients.

Endothelin receptor antagonist (ETRA) showed the promising result to prevent CVS but failed to show a benefit in term of mortality and neurological outcomes (Kramer and Fletcher, 2009; Ma et al., 2012; Macdonald et al., 2011; Vergouwen et al., 2012), which suggests that additional factors contribute to clinical deterioration and DCI, the main predictor of outcome. This strongly challenges the concept that angiographic vasospasm is the predominant factor for DCI. In particular, endothelin receptor antagonist was given in 5 trials with 2601 patients (Vergouwen et al., 2012). It showed an increased poor functional outcome despite CVS reduction; no effect was noticed regarding DCI, cerebral infarction, or case fatality rate. This emphasizes the various etiologies of DCI rather than to be narrowed down to CVS.

3. Interleukin-6

Interleukin-6 is a pleiotropic cytokine that plays a role in vascular and metabolic diseases (Hunter and Jones, 2015). IL6R(CD126), gp130(CD130), GTPase Ras-Raf, mitogen-activated protein kinases (MAPK), and Janus kinases (JAKs) signal transducer and activator of transcription constitute the signaling pathway of this cytokine with hormone-like activity (Hunter and Jones, 2015). IL-6 pro-inflammatory and anti-inflammatory property depending on the pathogenesis involved (Hunter and Jones, 2015). In aSAH, il-6 has a neuroinflammatory response (Suzuki et al., 2009).

The proteolytic processing of the IL-6 receptor on neutrophils turns on the signaling pathway in residing cells such as cerebral vasculature cells (Jones, 2005). IL-6 levels were positively linked to poor outcomes according to several studies (Muroi et al., 2013; Höllig, Remmel et al., 2015; Höllig, Thiel et al., 2015; Kao et al., 2015). We mention a detailed study that involved 80 aSAH patients. IL-6 serum levels were significantly raised in patients presenting with low Hunt & Hess (H&H) grade and patients with poor clinical outcome (Glasgow outcome scale 1-3), reflecting the inflammatory component of severe aSAH (Chaudhry et al., 2017). In a multivariate setting, serum IL-6 was independently associated with poor outcome assessed by mRS (modified Rankin Scale). More elevation in IL-6 serum level was noted in patients with concomitant intraventricular/intracranial bleed, hydrocephalus, and seizure (Chaudhry et al., 2017).

The role of inflammatory pathways in developing cerebral vasospasm and Delayed Ischemic Neurologic Deficit (DIND) was reflected with higher IL-6 level in the serum of these patients on day 3, 7 and 9, with levels approaching 1.5 ± 0.2 (log IL-6 pg/mL), compared with patients without DIND or vasospasm with serum IL-6 level 1.0 ± 0.2 (log IL-6 pg/mL) (Chaudhry et al., 2017).

Another study involved 24 patients with aSAH, where IL-6 serum levels were compared according to different intracranial pressure post-bleed. IL-6 levels were higher in patients with high intracranial pressure 5-9 days after SAH, indicating a severe inflammatory reaction. It was also correlated with low GOS levels, especially on 4-9 days post-aSAH (Graetz et al., 2010). Similar results were also found in another study, where CSF, but not plasma IL-6 levels, were positively correlated with DCI development, with concomitant C-reactive protein response. CSF levels were higher than plasma levels, which may be related to the smooth muscle production of IL-6, which reflects the vasospasm ensuing after aSAH (Sarrafzadeh et al., 2010).

Interventions by pharmacotherapy for IL-6 signaling interruptions reversed vasospasm in femoral artery vasospasm in a rat model (Bowman et al., 2004). This can bring us closer to achieve a better understanding of IL-6 role in the severity of aSAH and developing subsequent targeted therapy to reduce the complications.

In contrast to neuroinflammatory activation, immunodepression was introduced in various neurological conditions such as stroke and neurotrauma. (Woiciechowsky et al., 1998; Prass et al., 2003)Immunodepression was evident in aSAH. There were significantly lower counts of CD4 T cells, CD8 T cells, and natural killer cells as early as day 2 in symptomatic aSAH than asymptomatic aSAH. This has led to more infection rates -particularly pneumonia- in patients with symptomatic aSAH (Sarrafzadeh et al., 2010). This is important to mention since proteomics biomarkers can be minimal in these cases, requiring the implementation of preventive measures to avoid infections post aSAH.

4. Nesfatin-1

Nesfatin-1 is an anorexigenic peptide, combining calcium and Nucelobindin2, localized in the CSF and paraventricular nucleus of the hypothalamus (Wei et al., 2018). A study included 97 patients with aSAH. Those with poor outcomes and higher H&H grades had higher levels of nesfatin-1 than patients with good outcomes (11.342 ± 1.826 vs. 9.118 ± 0.278 ng/mL). Also, patients with DCI had higher levels of nesfatin-1 than patients with non-DCI (11.736 ± 1.844 vs. 8.842 ± 2.629 ng/mL) (Luo et al., 2019). Another study found that when 20 g/kg nesfatin-1 given to rats with induced SAH, they had better outcomes concerning neurological impairment and oxidative brain injury (Tang et al., 2012). On the other hand, there was an emphasis on the neuroprotective property of nesfatin-1 when injected in rats with subarachnoid hemorrhage-induced brain damage (Özsavcí et al., 2011). This seemingly contradictory result can be explained by the vasoconstrictive effect of nesfatin-1 as a defense mechanism against aneurysms or a rupture stimulant, as well as having an anti-apoptotic and anti-inflammatory effect (Cakir et al., 2017).

5. Thioredoxin (TRX)

TRX is a 12.5 kDa ubiquitous intracellular thiol protein involved in redox reactions. It modulates inflammation via its antioxidant property, in addition to its implication in cell growth and apoptosis (Koháryová and Kollárová, 2015). TRX has recently been considered a surrogate marker for severity and poor outcome in intracerebral hemorrhage, head trauma and ischemic strokes (Qi et al., 2015; Pan et al., 2016; Qian et al., 2016). A study found a significant elevation of TRX in 132 patients with aSAH (23.4 ± 12.2 ng/mL), compared with 132 controls (8.5 ± 4.0 ng/mL) association between serum TRX levels and modified Fischer score upon admission and 6 months’ predictor of mortality and unfavorable outcome. The optimal cut-off value of serum Trx levels as a predictor for 6-month mortality was considered to be 25.1 ng/mL (Dai et al., 2016). TRX levels improved the predictive performance of GCS score for 6 months’ outcome with a cutoff value of 20.3 ng/mL, but not for 1-week mortality or 6 months mortality when 112 patients with aSAH and 112 controls were studied (Pan et al., 2016).

TRX has reflected the oxidative stress that occurs during subarachnoid hemorrhage. Both studies mentioned above had well correlated with prediction of the outcome when TRX was taken upon admission. It constitutes an excellent predictive value in terms of outcome, yet no data have been mentioned regarding vasospasm and DCI prediction.

6. Creactive protein

CRP is an inflammatory marker of the pentraxin family, well known for its sensitivity. Its synthesis in hepatocytes highly depends on IL-6 and IL-1 (Mazlam and Hodgson, 1994). Many studies have demonstrated that an elevated CRP level can predict cerebral vasospasm and, subsequently, DIND (Fountas et al., 2009). Similarly, patients who presented with high Fischer and H&H grades on admission have developed higher CRP levels in serum (6-7 mg/L) (Romero et al., 2014).

The more unsatisfactory outcome expressed in GOS was correlated with higher serum CRP levels, which peaked on the 3 r d and 4 t h days post aSAH (8-10 mg/L) (Romero et al., 2014). Patients with clinical vasospasm had CRP levels reaching 16 mg/L (Romero et al., 2014).

CRP findings in aSAH correspond to the possible prediction of the clinical outcome. It reflects the severity of aSAH by being a surrogate of inflammation. Yet, it is confounded that many of these patients had concomitant systemic infections or other conditions that could elevate CRP levels. CRP will not be a specific marker during aSAH, although its sensitivity is well proved.

7. Copeptin

A protein released as an equimolar ratio with vasopressin from the same precursor protein, preprovasopressin, consists of 164-amino acid (Morgenthaler et al., 2006). Since vasopressin is involved in the potentiation of corticotrophin-releasing hormone, which increases Adrenocorticotropic hormone, it leads to cortisol production. Hence, Copeptin -the surrogate marker for vasopressin release- is considered a stress response marker (Morgenthaler et al., 2006). Many studies chose Copeptin due to its stability that remains intact for days, thus allowing easy blood sample measurement (Morgenthaler et al., 2006).

Median copeptin levels in healthy individuals have been reported and range from 3.7 to 4.2 pmol/L (Katan et al., 2009). A study found a significant association between higher copeptin level and WFNS grade 4 and 5 with a median of 26.3 pmol/L, Fischer grade 3 with 18.3 pmol/L, and ICH (intracerebral hemorrhage) with 52.5 pmol/L, along with an association with the amount of subarachnoid blood. Nevertheless, it has been concluded that admission copeptin levels are not reliable for predicting vasospasm or delayed cerebral ischemia (Fung et al., 2013).

Zhu et al. (2011) and colleagues reported a correlation between the elevated serum copeptin levels with vasospasm during the subacute period and increased mortality at 1 year, but should not be used since clinical grades were found to have higher accuracy.

8. Albumin

Albumin is a negative acute phase reactant, known to fall acutely due to metabolic response to injury. Low nutritional status is well known for critical illness and unfavorable prognosis (McCluskey et al., 1996). Considering albumin, a neuroprotective protein, Kapoor and colleagues found that patients having lower albumin ( < 3.5 mg/dL) levels at admission had higher rates of neurological deficits, infarcts, mortality, unfavorable GOS, H&H grade and Fisher grade. Also, at 1 week, a fall in albumin level compared to the admission level was significantly linked to unfavorable GOS and mortality (Kapoor et al., 2018).

Different trials were conducted and showed a neuroprotective effect of albumin when given after subarachnoid hemorrhage as a promising tool to reduce morbidity and mortality (Wang et al., 2017; Suarez et al., 2012).

9. Myeloperoxidase (MPO)

MPO is an essential enzyme in the neutrophilic respiratory burst response to pathogens found in leukocytes’ lysosomes (Lim et al., 2012). As a marker of inflammation increased, MPO was associated with poor prognosis in patients with aSAH. Lim et al. (2012) and colleagues noted that patients with symptomatic vasospasm compared to those without vasospasm had MPO levels of 115.5 ng/mL vs. 59.4 ng/mL respectively, 2 or more days before the vasospastic event.

Again, inflammation was highly suggested as the core cause of complications following subarachnoid hemorrhage.

10. Neuron-specific enolase (NSE)

NSE has been used as a biomarker for neuronal damage, and it is a glycolytic enzyme mainly localized in the neuron (Butterworth et al., 1996). Mabe and colleagues found that NSE levels in serum were significantly higher in patients with aSAH (29 patients) than patients without a normal range of 3.1 ± 0.6 ng/mL in serum. Also, the serum level increased with higher Fischer grades. Grades 2, 3, and 4 were 3.6 ± 1.3, 5.6 ± 2.1, and 7.6 ± 1.2 ng/mL, respectively, when taken within 3 days of aSAH (Mabe et al., 1991).

Kacira et al. (2007) and colleagues demonstrated an increasing level of serum NSE towards day 7 in 20 patients having aSAH, with values reaching 18.54 ± 19.86 μ g/L compared with 5.94 ± 2.20 μ g/L in a control group, and this was attributed to increased neuronal damage. Furthermore, Tawk et al. (2016) and colleagues involved 309 patients. They found that NSE level > 15 ng/mL is considered relatively high and significantly associated with higher WFNS score, GCS, and H&H grade, concluding that NSE can be a potential predictor for poor outcome in aSAH. Prognostic studies can be based on the cutoff values found to incriminate this level officially in the clinical routine.

11. S100B

S100B is a calcium-binding peptide. Its production is carried out in astrocytes and has powerful paracrine and autocrine effects on neurons and glia (Rothermundt et al., 2003). It regulates the cytoskeletal structure and cell proliferation. At nanomolar concentrations, S100B improves neuronal survival and outgrowth in the cerebral cortex and decreases mitochondrial function loss during glucose shortage (Weiss et al., 2006).

In 55 patients with SAH, the mean daily value of S100B above 0.4 μ g/L was higher in patients with high WFNS and Fisher scores (Weiss et al., 2006). S100B prognostic review of 74 patients with SAH also showed a significant correlation between high serum S100B (mean 0.17 μ g/L) and poor prognosis as measured by GOS (Glasgow outcome scale), as well as cerebral infarction following aSAH (Moritz et al., 2010). Neither NSE nor S100B was correlated with the development of cerebral vasospasm (Moritz et al., 2010).

12. Conclusions

Aneurysmal Subarachnoid hemorrhage prognosis depends on the debilitating secondary outcome. Ongoing research to improve clinical management have surfaced to light in the past few years, mostly when numerous proteomics are studied to improve their predictive value regarding CVS and DCI after aSAH. Understanding the disease pathogenesis can open the opportunity to decipher how this proteomics relates to aSAH and thus translating them into clinical routine. Monitoring these biomarkers’ trends allows the classification of patients in terms of poor outcome even before their neurological deterioration, and the prediction of various complications, hence identifying patients who need close monitoring and aggressive management.

Proteomics helped unravel many pathophysiologic processes, allowing medical interferences along several pathways to be used as future targeted therapy. Proteomics in the serum may help us better fathom the systemic response with its repercussions on the brain’s local processes. A prospective clinical trial should be conducted to evaluate these proteomics’ effectiveness, sensitivity and specificity, biokinetics, specific peaks, and half-lives. So far, not a single proteomic biomarker has proven to have sufficient sensitivity or specificity for DCI’s reliable prediction. More proteomics should be complementary to concomitant neuroimaging and neuromonitoring techniques rather than replacing current modalities. DCI’s complex pathophysiology makes it complicated to set a definitive pattern of biomarkers that can be a potential target for preventing complications.

Abbreviations

aSAH, Aneurysmal subarachnoid hemorrhage; CRP: C Reactive Protein; CVS: Cerebral Vasospasm; CSF: Cerebrospinal Fluid; DCI: Delayed Cerebral Ischemia; DIND: Delayed Ischemic Neurologic Deficit; ET-1: Endothelin-1; GOS: Glasgow Outcome scale; NSE: Neuron-specific enolase; MPO: Myeloperoxidase; TRX: Thioredoxin; WFNS: World Federation of Neurological surgeons.

Author contributions

Shadi and Hani wrote the paper; Safwan, Charbel and Mohamad gathered the articles and did the tables. Dr Tarek Sunna reviewed the paper and edited it.

Acknowledgment

Thanks to all the peer reviewers and editors for their opinions and suggestions.

Conflict of Interest

The authors declare no competing interests.

References
[1]
Bowman, G., Dixit, S., Bonneau, R. H., Chinchilli, V. M. and Cockroft, K. M. (2004) Neutralizing Antibody against Interleukin-6 Attenuates Posthemorrhagic Vasospasm in the Rat Femoral Artery Model. Neurosurgery 54, 719-726.
[2]
Butterworth, R. J., Wassif, W. S., Sherwood, R. A., Gerges, A., Poyser, K. H., Garthwaite, J., Peters, T. J. and Bath, P. M. (1996) Serum neuron-specific enolase, carnosinase, and their ratio in acute stroke. An enzymatic test for predicting outcome? Stroke 27, 2064-2068.
[3]
Cakir, M., Calikoglu, C., Yılmaz, A., Akpinar, E., Bayraktutan, Z. and Topcu, A. (2017) Serum nesfatin-1 levels: a potential new biomarker in patients with subarachnoid hemorrhage. International Journal of Neuroscience 127, 154-160.
[4]
Chaudhry, S., Stoffel-Wagner, B., Kinfe, T., Güresir, E., Vatter, H., Dietrich, D., Lamprecht, A. and Muhammad, S. (2017) Elevated systemic IL-6 levels in patients with aneurysmal subarachnoid hemorrhage is an unspecific marker for post-SAH complications. International Journal of Molecular Sciences 18, 2580.
[5]
Dai, J., Cai, J., Lin, Q., Chen, X., Lu, C., Sun, J. and Ba, H. (2016) Thioredoxin as a marker for severity and prognosis of aneurysmal subarachnoid hemorrhage. Journal of the Neurological Sciences 363, 84-89.
[6]
De Marchis, G. M., Filippi, C. G., Guo, X., Pugin, D., Gaffney, C. D., Dangayach, N. S., Suwatcharangkoon, S., Falo, M. C., Schmidt, J. M., Agarwal, S., Connolly, E. S., Claassen, J., Zhao, B. and Mayer, S. A. (2015) Brain injury visible on early MRI after subarachnoid hemorrhage might predict neurological impairment and functional outcome. Neurocritical Care 22, 74-81.
[7]
Dreier, J. P. (2011) The role of spreading depression, spreading depolarization and spreading ischemia in neurological disease. Nature Medicine 17, 439-447.
[8]
Eriksen, N., Rostrup, E., Fabricius, M., Scheel, M., Major, S., Winkler, M. K. L., Bohner, G., Santos, E., Sakowitz, O. W., Kola, V., Reiffurth, C., Hartings, J. A., Vajkoczy, P., Woitzik, J., Martus, P., Lauritzen, M., Pakkenberg, B. and Dreier, J. P. (2019) Early focal brain injury after subarachnoid hemorrhage correlates with spreading depolarizations. Neurology 92, e326-e341.
[9]
Fountas, K. N., Tasiou, A., Kapsalaki, E. Z., Paterakis, K. N., Grigorian, A. A., Lee, G. P. and Robinson, J. S. (2009) Serum and cerebrospinal fluid C-reactive protein levels as predictors of vasospasm in aneurysmal subarachnoid hemorrhage. Neurosurgical Focus 26, E22.
[10]
Frontera, J. A., Fernandez, A., Schmidt, J. M., Claassen, J., Wartenberg, K. E., Badjatia, N., Parra, A., Connolly, E. S. and Mayer, S. A. (2008) Impact of nosocomial infectious complications after subarachnoid hemorrhage. Neurosurgery 62, 80-87.
[11]
Fung, C., De Marchis, G. M., Katan, M., Seiler, M., Arnold, M., Gralla, J., Raabe, A. and Beck, J. (2013) Copeptin as a marker for severity and prognosis of aneurysmal subarachnoid hemorrhage. PLoS One 8, e53191.
[12]
Gewirtz, R. J., Dhillon, H. S., Goes, S. E., DeAtley, S. M. and Scheff, S. W. (1999) Lactate and free fatty acids after subarachnoid hemorrhage. Brain Research 840, 84-91.
[13]
Graetz, D., Nagel, A., Schlenk, F., Sakowitz, O., Vajkoczy, P. and Sarrafzadeh, A. (2010) High ICP as trigger of proinflammatory IL-6 cytokine activation in aneurysmal subarachnoid hemorrhage. Neurological Research 32, 728-735.
[14]
Haley, E. C., Kassell, N. F. and Torner, J. C. (1992) The international cooperative study on the timing of aneurysm surgery. The North American experience. Stroke 23, 205-214.
[15]
Hartings, J. A., York, J., Carroll, C. P., Hinzman, J. M., Mahoney, E., Krueger, B., Winkler, M. K. L., Major, S., Horst, V., Jahnke, P., Woitzik, J., Kola, V., Du, Y., Hagen, M., Jiang, J. and Dreier, J. P. (2017) Subarachnoid blood acutely induces spreading depolarizations and early cortical infarction. Brain 140, 2673-2690.
[16]
Höllig, A., Remmel, D., Stoffel-Wagner, B., Schubert, G. A., Coburn, M. and Clusmann, H. (2015) Association of early inflammatory parameters after subarachnoid hemorrhage with functional outcome: a prospective cohort study. Clinical Neurology and Neurosurgery 138, 177-183.
[17]
Höllig, A., Thiel, M., Stoffel-Wagner, B., Coburn, M. and Clusmann, H. (2015) Neuroprotective properties of dehydroepiandrosterone-sulfate and its relationship to interleukin 6 after aneurysmal subarachnoid hemorrhage: a prospective cohort study. Critical Care 19, 231.
[18]
Hunt, W. E. and Hess, R. M. (1968) Surgical risk as related to time of intervention in the repair of intracranial aneurysms. Journal of Neurosurgery 28, 14-20.
[19]
Hunter, C. A. and Jones, S. A. (2015) IL-6 as a keystone cytokine in health and disease. Nature Immunology 16, 448-457.
[20]
Jean Beltran, P. M., Federspiel, J. D., Sheng, X. and Cristea, I. M. (2017) Proteomics and integrative omic approaches for understanding host–pathogen interactions and infectious diseases. Molecular Systems Biology 13, 922.
[21]
Jones, S. A. (2005) Directing transition from innate to acquired immunity: defining a role for IL-6. Journal of Immunology 175, 3463-3468.
[22]
Juvela, S. (2000) Plasma endothelin concentrations after aneurysmal subarachnoid hemorrhage. Journal of Neurosurgery 92, 390-400.
[23]
Kacira, T., Kemerdere, R., Atukeren, P., Hanimoglu, H., Sanus, G. Z., Kucur, M., Tanriverdi, T., Gumustas, K. and Kaynar, M. Y. (2007). Detection of caspase-3, neuron specific enolase, and high-sensitivity C-reactive protein levels in both cerebrospinal fluid and serum of patients after aneurysmal subarachnoid hemorrhage. Neurosurgery 60, 674-680.
[24]
Kao, H. W., Lee, K. W., Kuo, C. L., Huang, C. S., Tseng, W. M., Liu, C. S. and Lin, C. P. (2015) Interleukin-6 as a prognostic biomarker in ruptured intracranial aneurysms. PLoS One 10, e0132115.
[25]
Kapoor, A., Dhandapani, S., Gaudihalli, S., Dhandapani, M., Singh, H. and Mukherjee, K. K. (2018) Serum albumin level in spontaneous subarachnoid hemorrhage: more than a mere nutritional marker! British Journal of Neurosurgery 32, 47-52.
[26]
Katan, M., Fluri, F., Morgenthaler, N. G., Schuetz, P., Zweifel, C., Bingisser, R., Müller, K., Meckel, S., Gass, A., Kappos, L., Steck, A. J., Engelter, S. T., Müller, B. and Christ-Crain, M. (2009) Copeptin: a novel, independent prognostic marker in patients with ischemic stroke. Annals of Neurology 66, 799-808.
[27]
Koháryová, M. and Kollárová, M. (2015). Thioredoxin system - a novel therapeutic target. General Physiology and Biophysics 34, 221-233.
[28]
Kramer, A. and Fletcher, J. (2009) Do endothelin-receptor antagonists prevent delayed neurological deficits and poor outcomes after aneurysmal subarachnoid hemorrhage? Stroke 40, 3403-3406.
[29]
Lim, M., Bower, R. S., Wang, Y., Sims, L., Bower, M. R., Camara-Quintana, J., Li, G., Cheshier, S., Harsh, G. R., Steinberg, G. K. and Guccione, S. (2012) The predictive value of serum myeloperoxidase for vasospasm in patients with aneurysmal subarachnoid hemorrhage. Neurosurgical Review 35, 413-419.
[30]
Lovelock, C. E., Rinkel, G. J. E. and Rothwell, P. M. (2010) Time trends in outcome of subarachnoid hemorrhage: Population-based study and systematic review. Neurology 74, 1494-1501.
[31]
Lublinsky, S., Major, S., Kola, V., Horst, V., Santos, E., Platz, J., Sakowitz, O., Scheel, M., Dohmen, C., Graf, R., Vatter, H., Wolf, S., Vajkoczy, P., Shelef, I., Woitzik, J., Martus, P., Dreier, J. P. and Friedman, A. (2019) Early blood-brain barrier dysfunction predicts neurological outcome following aneurysmal subarachnoid hemorrhage. Ebiomedicine 43, 460-472.
[32]
Lückl, J., Lemale, C. L., Kola, V., Horst, V., Khojasteh, U., Oliveira-Ferreira, A. I., Major, S., Winkler, M. K. L., Kang, E., Schoknecht, K., Martus, P., Hartings, J. A., Woitzik, J. and Dreier, J. P. (2018) The negative ultraslow potential, electrophysiological correlate of infarction in the human cortex. Brain 141, 1734-1752.
[33]
Luo, W., Chen, D., Wang, H. and Hu, J. (2019) An elevated serum level of nesfatin-1 may predict poor outcomes in patients with aneurysmal subarachnoid hemorrhages. International Journal of Clinical and Experimental Medicine 12, 5744-5749.
[34]
Ma, J., Huang, S., Ma, L., Liu, Y., Li, H. and You, C. (2012) Endothelin-receptor antagonists for aneurysmal subarachnoid hemorrhage: an updated meta-analysis of randomized controlled trials. Critical Care 16, R198.
[35]
Mabe, H., Suzuki, S., Mase, M., Umemura, A. and Nagai, H. (1991) Serum neuron-specific enolase levels after subarachnoid hemorrhage. Surgical Neurology 36, 170-174.
[36]
Macdonald, R. L., Higashida, R. T., Keller, E., Mayer, S. A., Molyneux, A., Raabe, A., Vajkoczy, P., Wanke, I., Bach, D., Frey, A., Marr, A., Roux, S. and Kassell, N. (2011) 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). The Lancet Neurology 10, 618-625.
[37]
Mazlam, M. Z. and Hodgson, H. J. (1994) Interrelations between interleukin-6, interleukin-1 beta, plasma C-reactive protein values, and in vitro C-reactive protein generation in patients with inflammatory bowel disease. Gut 35, 77-83.
[38]
McCluskey, A., Thomas, A. N., Bowles, B. J. M. and Kishen, R. (1996) The prognostic value of serial measurements of serum albumin concentration in patients admitted to an intensive care unit. Anaesthesia 51, 724-727.
[39]
Minhas, P. S., Menon, D. K., Smielewski, P., Czosnyka, M., Kirkpatrick, P. J., Clark, J. C. and Pickard, J. D. (2003) Positron emission tomographic cerebral perfusion disturbances and transcranial doppler findings among patients with neurological deterioration after subarachnoid hemorrhage. Neurosurgery 52, 1017-1024.
[40]
Morgenthaler, N. G., Struck, J., Alonso, C. and Bergmann, A. (2006) Assay for the measurement of copeptin, a peptide derived from the precursor of vasopressin. Clinical Chemistry 52, 112-119.
[41]
Moritz, S., Warnat, J., Bele, S., Graf, B. M. and Woertgen, C. (2010) The prognostic value of NSE and S100B from serum and cerebrospinal fluid in patients with spontaneous subarachnoid hemorrhage. Journal of Neurosurgical Anesthesiology 22, 21-31.
[42]
Mrozek, S., Dumurgier, J., Citerio, G., Mebazaa, A. and Geeraerts, T. (2014) Biomarkers and acute brain injuries: interest and limits. Critical Care 18, 220.
[43]
Muroi, C., Hugelshofer, M., Seule, M., Tastan, I., Fujioka, M., Mishima, K. and Keller, E. (2013) Correlation among systemic inflammatory parameter, occurrence of delayed neurological deficits, and outcome after aneurysmal subarachnoid hemorrhage. Neurosurgery 72, 367-375.
[44]
Özsavcí, D., Erşahin, M., Şener, A., Özakpinar, Ö. B., Toklu, H. Z., Akakín, D., Şener, G. and Yeğen, B. Ç. (2011) The novel function of nesfatin-1 as an anti-inflammatory and antiapoptotic peptide in subarachnoid haemorrhage-induced oxidative brain damage in rats. Neurosurgery 68, 1699-1708.
[45]
Pan, D., Le, H., Yan, M., Hassan, M., Gong, J. and Wang, H. (2016) Change of serum levels of thioredoxin in patients with severe traumatic brain injury. Clinica Chimica Acta 453, 62-66.
[46]
Pandey, A. and Mann, M. (2000) Proteomics to study genes and genomes. Nature 405, 837-846.
[47]
Prass, K., Meisel, C., Höflich, C., Braun, J., Halle, E., Wolf, T., Ruscher, K., Victorov, I. V., Priller, J., Dirnagl, U., Volk, H. D. and Meisel, A.(2003) Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. The Journal of Experimental Medicine 198, 725-736.
[48]
Qi, A., Li, Y., Liu, Q., Si, J., Tang, X., Zhang, Z., Qi, Q. and Chen, W. (2015) Thioredoxin is a novel diagnostic and prognostic marker in patients with ischemic stroke. Free Radical Biology and Medicine 80, 129-135.
[49]
Qian, S., Hu, X., He, S., Li, B., Zheng, X. and Pan, G. (2016) Prognostic value of serum thioredoxin concentrations after intracerebral hemorrhage. Clinica Chimica Acta 455, 15-19.
[50]
Romero, F. R., Cataneo, D. C. and Cataneo, A. J. M. (2014) C-reactive protein and vasospasm after aneurysmal subarachnoid hemorrhage1. Acta Cirurgica Brasileira 29, 340-345.
[51]
Rothermundt, M., Peters, M., Prehn, J. H. M. and Arolt, V. (2003) S100B in brain damage and neurodegeneration. Microscopy Research and Technique 60, 614-632.
[52]
Sarrafzadeh, A., Schlenk, F., Gericke, C. and Vajkoczy, P. (2010) Relevance of Cerebral Interleukin-6 after aneurysmal subarachnoid hemorrhage. Neurocritical Care 13, 339-346.
[53]
Seibert, V. (2005) Advances in clinical cancer proteomics: SELDI-ToF-mass spectrometry and biomarker discovery. Briefings in Functional Genomics and Proteomics 4, 16-26.
[54]
Shevchenko, A., Zachariae, W. and Shevchenko, A. (1999) A strategy for the characterization of protein interaction networks by mass spectrometry. Biochemical Society Transactions 27, 549-554.
[55]
Suarez, J. I., Martin, R. H., Calvillo, E., Dillon, C., Bershad, E. M., MacDonald, R. L., Wong, J. and Harbaugh, R. (2012) The albumin in subarachnoid hemorrhage (ALISAH) multicenter pilot clinical trial. Stroke 43, 683-690.
[56]
Suhardja, A. (2004) Mechanisms of disease: roles of nitric oxide and endothelin-1 in delayed cerebral vasospasm produced by aneurysmal subarachnoid hemorrhage. Nature Clinical Practice Cardiovascular Medicine 1, 110-116.
[57]
Suzuki, S., Tanaka, K. and Suzuki, N. (2009) Ambivalent aspects of interleukin-6 in cerebral ischemia: Inflammatory versus neurotrophic aspects. Journal of Cerebral Blood Flow Metabolism 29, 464-479.
[58]
Tang, C., Fu, X., Xu, X., Wei, X. and Pan, H. (2012) The anti-inflammatory and anti-apoptotic effects of nesfatin-1 in the traumatic rat brain. Peptides 36, 39-45.
[59]
Tawk, R. G., Grewal, S. S., Heckman, M. G., Rawal, B., Miller, D. A., Edmonston, D., Ferguson, J. L., Navarro, R., Ng, L., Brown, B. L., Meschia, J. F. and Freeman, W. D. (2016) The relationship between serum neuron-specific enolase levels and severity of bleeding and functional outcomes in patients with nontraumatic subarachnoid hemorrhage. Neurosurgery 78, 487-491.
[60]
Vergouwen, M. D. I., Algra, A. and Rinkel, G. J. E. (2012) Endothelin receptor antagonists for aneurysmal subarachnoid hemorrhage. Stroke 43, 2671-2676.
[61]
Wang, L., Li, M., Xie, Y., Xu, L., Ye, R. and Liu, X. (2017) Preclinical efficacy of human Albumin in subarachnoid hemorrhage. Neuroscience 344, 255-264.
[62]
Wei, Y., Li, J., Wang, H. and Wang, G. (2018) NUCB2/nesfatin-1: expression and functions in the regulation of emotion and stress. Progress in Neuro-Psychopharmacology and Biological Psychiatry 81, 221-227.
[63]
Weiss, N., Sanchez-Peña, P., Roche, S., Beaudeux, J., Colonne, C., Coriat, P. and Puybasset, L. (2006) Prognosis value of plasma S100B protein levels after subarachnoid aneurysmal hemorrhage. Anesthesiology 104, 658-666.
[64]
Wilkins, M. R., Sanchez, J., Gooley, A. A., Appel, R. D., Humphery-Smith, I., Hochstrasser, D. F. and Williams, K. L. (1996) Progress with proteome projects: why all proteins expressed by a genome should be identified and how to do it. Biotechnology and Genetic Engineering Reviews 13, 19-50.
[65]
Woiciechowsky, C., Asadullah, K., Nestler, D., Eberhardt, B., Platzer, C., Schöning, B., Glöckner, F., Lanksch, W. R., Volk, H. D., and Döcke, W. D. (1998) Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by brain injury. Nature Medicine 4, 808-813.
[66]
Zhu, X., Chen, J., Zhou, F., Liu, Q., Chen, G. and Zhang, J. (2011) Detection of copeptin in peripheral blood of patients with aneurysmal subarachnoid hemorrhage. Critical Care 15, R288.
Share
Back to top