· Login       · Register
View
go to the main page
Articles & issues
sns-share   facebook go Twitter go Google+ go
Review article
Neuroprotective Agents in the Intensive Care Unit
- Neuroprotective Agents in ICU -
Yunes Panahi 1,2, Mojtaba Mojtahedzadeh 2,3, Atabak Najafi 4, Seyyed Mahdi Rajaee 4, Mohammad Torkaman 5, Amirhossein Sahebkar 6,7,8 *
1 Clinical Pharmacy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
2 Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran, Iran
3 Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
4 Gastrointestinal Pharmacology Interest Group(GPIG), Universal Scientific Education and Research Network(USERN), Tehran, Iran
5 Department of Pediatrics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
6 Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
7 Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
8 School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
* Amirhossein Sahebkar. Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran. Tel: +98-511-800-2288 Fax: +98-511-800-2287 E-mail: sahebkara@mums.ac.ir; amir_saheb2000@yahoo.com
[received date: 2017-12-21 / accepted date: 2018-11-14]
Abstract
Neuroprotection or prevention of neuronal loss is a complicated molecular process that is mediated by various cellular pathways. Use of different pharmacological agents as neuroprotectants has been reported especially in the last decades. These neuroprotective agents act through inhibition of inflammatory processes and apoptosis, attenuation of oxidative stress and reduction of free radicals. Control of this injurious molecular process is essential to the reduction of neuronal injuries and is associated with improved functional outcomes and recovery of the patients admitted to the intensive care unit. This study reviews neuroprotective agents and their mechanisms of action against central nervous system damages.
Keywords
neuroprotection, intensive care unit, stroke
Open Access
This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Neuroprotection aims at preventing neuronal loss and Received: Dec 21, 2017 Reviewed: Aug 09, 2018 Accepted: Nov 14, 2018 neurodegeneration through applying different agents to inhibit pathophysiological pathways that are injurious to the nervous system [1]. Use of neuroprotective agents has a long history from ancient Greece to the current age with the presence of pharmacological and natural neuroprotectants and gene therapies [1]. The most common conditions associated with nervous system involvement and Intensive Care Unit (ICU) admission are Trauma, shock, stroke, sepsis, traumatic brain injury (TBI) and ruptured brain aneurysm [1, 2].

2. Stroke
Stroke is one of the major causes of disability and death in the world [3]. Only one-third of patients with stroke recover enough to be free of disability [4]. Males have a greater incidence of stroke than females; hypertension, diabetes, atrial fibrillation, smoking and oral contraception pills are the major risk factors for stroke [2]. After stroke, decreased blood flow and subsequent disturbance in ionic homeostasis and intracellular edema are major consequences. Release of excitatory neurotransmitters and production of free radicals because of mitochondrial dysfunction occur consequently [5]. Oxidative stress, activation of apoptotic pathways and excitotoxicity are the subsequent events after cerebral ischemia that lead to neuronal death [4, 5].

3. Shock
Shock is defined as a decrease in blood perfusion to the body tissues and consequent deficiency in oxygen and substrate due to tissues and cell injuries [6]. Main types of shock include cardiogenic, hypovolemic, anaphylactic, septic and neurogenic shock [7]. Cellular ischemia is the primary cause of cell damage. After a decrease in blood perfusion to the cells, aerobic generation of ATP will decrease and mitochondrial dysfunction, increased intracellular PH, production of free radicals and autolytic pathway activation are following findings [6, 7].

4. Sepsis
Sepsis is a fatal condition with a high mortality rate [8]. Severe sepsis can lead to hypoperfusion and subsequent increase in serum creatinine level, an increase of serum lactate and total bilirubin level, thrombocytopenia and acute lung injuries [9]. Pneumonia, urinary tract infection and intra-abdominal infections are the most common causes of sepsis [10]. The incidence of Gram-negative bacterial infections have increased during the past decade [11]. The proinflammatory and anti-inflammatory responses are implicated in the tissue damage and secondary bacterial infection but specific responses depend on the host immune system and the causative pathogen [10, 11].

5. Traumatic brain injury
Trauma is one of the primary causes of disability and death worldwide [12]. TBI occurs as a result of sudden trauma to the head and leads to cognition, motor function and sensation impairment with a high mortality rate [13]. Increase in intracranial pressure, focal contusion, hematoma and cerebral edema formation occurs after trauma to the head [14]. The secondary part of TBIs will occur in cellular stage with severe consequences that have been described by Park and colleagues (2008) as; “(1) failure of neuronal energy, (2) glial injury and dysfunction, (3) inflammation, (4) destruction and stenosis of microvasculature, (5) excitotoxicity (6) and aberrant ionic homeostasis in neurons” [15].

6. Ruptured Brain Aneurysm
A brain aneurysm or a cerebral aneurysm is dilation of a supplied blood artery of the brain [16]. An unruptured aneurysm is often asymptomatic and recognizable by computed tomography or magnetic resonance imaging. The consequence of a ruptured brain aneurysm is subarachnoid hemorrhage that is a life-threatening condition [16]. Most of the brain aneurysms are congenital or with the familial background and genetic predisposition [17].

7. Mechanisms of neuronal injuries
Programmed cell dead (PCD), particularly apoptosis, excitotoxicity, oxidative stress, and inflammation are the primary mechanisms leading to neuronal injuries in the patients admitted to ICU [18]. PCD is a mixture of pathways that result in removal of the unwanted cell [19]. Several proteins such as caspases, apoptosis-inducing factor, Bcl-2 family proteins, p53 protein, tumor necrosis factor receptor, TRADD, Fas ligand and Fas-associated protein with death domain (FADD) are essential in the activation or inhibition of PCD pathways [19]. Three major routes have been defined for PCD that include intrinsic, extrinsic and caspase-independent pathways [20]. Some of the Bcl-2 family proteins have a significant role in the initiation of PCD by increasing mitochondrial permeability, the release of cytochrome-c from mitochondria and activation of caspases, known as the intrinsic pathway of apoptosis[21]. The extrinsic pathway works through triggering caspases-8 via membrane receptors like Fas and tumor necrosis factor-α [19, 20]. The release of apoptosis-inducing factor from mitochondria can induce apoptosis through a caspase-independent pathway [20]. Excitotoxicity is a primary mechanism of neuronal damage [22]. Glutamatergic neurons play an important role in the excitotoxicity [23]. As described by Dong et al. (2009), “glutamate can activate N-methyl-D-aspartic acid (NMDA), α-amino-3-hydroxy-5methylisoxazole-4-propionate (AMPA) and kainic acid (KA) receptors” [22]. Overactivation of glutamate receptors can lead to continuous activation of these receptors [24]. This constant activation results in: 1) cellular calcium hemostasis impairments [25], 2) oxidative stress and increased production of nitric oxide [26, 27], 3) generation of free radicals such as peroxynitrite and hydroxyl radical [28], and 4) PCD. Oxidative stress is associated with the accumulation of reactive species of nitrogen and oxygen (ROS) in the cell that leads to ATP depletion, mitochondrial dysfunction and impairment of cellular hemostasis [29]. Free radicals such as hydroxyl are highly active and can pair with DNA and lead to oxidation of DNA and cell death [29]. Oxidative stress and production of free radicals result in the activation of PCD [30]. In the ischemic neuronal injuries, the presence of inflammatory cytokines, leukocytes, and chemokines play a significant role in the pathogenesis of cell death [31, 32]. Cerebral blood flow occlusion leads to inflammatory reactions [33]. The presence of leukocytes and inflammatory mediators increase the level of adhesion proteins such as P-Selectin, E-Selectin and ICAM-1 results in the obstruction of microvessels, edema, necrosis and infarction [32, 33].

8. Neuroprotective agents
Beneficial use of many agents has been reported in the prevention of neuronal cell death in animal models but supportive data from clinical trials is still lacking [34]. New drugs have been introduced during the last decade with better outcomes in patients. We discuss some potent neuroprotective agents that may be beneficial for patients admitted to the ICU (Table 1).

Table. Table 1.1
Table 1.1


Table. Table 1.2
Table 1.2


  • Potential neuroprotective agent in the intensive care unit for the management of hemorrhagic stroke, ischemic stroke and traumatic brain injuries.
8.1. Glutamate blockers
Glutamate is a neurotrasmitter and as described by Danysz et al. (2002), “it activates three major types of iono tropic receptors, namelyo~-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainateand N-methyl-D-aspartate (NMDA) and several types of metabotropic receptors. AMPA receptors are involved in fast glutamatergic neurotransmission” [35]. The major role of glutamate blockers are inhibition of glutamate binding to NMDA and AMPA receptors to avoid excitotoxicity [36]. Glutamate blockers such as polyarginine R18 and NA-1 (TAT-NR2B9c) were used in different studies in stroke models in rats [37, 38], non-human primates [39] and also in human [40]. Milani and colleagues reported R18 as a potential neuroprotective agent. In both studies on rat administration of R18 in 60 minutes post-stroke reduced infarct volume and cerebral edema, improved functional outcome, more efficient than NA-1 [37, 38]. These agents have Anti-excitotoxicity properties with inhibition of postsynaptic density-95 protein/nNOS complex [41]. Reduce of oxidative stress of mitochondria in neurons [42], reduction of calcium influx due to glutamate excitotoxic [43], proteolytic activity inhibition of proprotein convertases [44], are mechanisms of action for this class of neuroprotective agents.

8.2. Magnesium sulfate
Magnesium is the second abundant cation in the body. It is involved in different physiological pathways and has different clinical applications [45-47]. Magnesium activates the enzymatic process for the transfer of phosphate from ADP to ATP. It regulates intracellular calcium availability, cell cycle and mitochondrial function. Decrease of serum magnesium levels lead to hypocalcemia and hypokalemia. Magnesium also blocks NMDA receptor and leads to analgesia and neuroprotection [48]. Magnesium sulfate (MgSO4) is another potentiate neuroprotective agent with anti-excitotoxicity activity, blockage of N-methyl-D-aspartate (NMDA) channels and voltage-gated calcium channels inhibition properties [49, 50]. Use of MgSO4 have been documented in acute stroke [51-60], aneurysmal subarachnoid hemorrhage [61-68], and Traumatic brain injuries [49, 50, 69]. Monitoring of magnesium is vital for patients admitted to the ICU because low serum magnesium level is associated with high mortality rate in the ICU [70, 71]. The use of MgSO4 in patients admitted to ICU has been associated with a decrease in the biomarkers such as S100B protein and serum neuron-specific enolase level (S-NSE) [72, 73].

8.3. Statins
Statins or 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors are the most frequntly administered class of cholesterol-lowering drugs [74] that have established effects in reducing coronary plaque volume [75] and cardiovascular events [76]. Beyond their wellknown hypocholesterolemic effect, a myriad of lipid-independent pleiotropic activities have been described for statins [77-88]. Statins need to enter the cells, some of them are lipophilic and some are hydrophilic. Statins have different intestinal absorption after oral administration, differing from 30% (lovastatin) to 98% (fluvastatin). Systemic bioavailability of these drugs are 5-30%. Most of them are metabolized by cytochromeP-450 and have high protein binding [89]. Statins have antioxidant and anti-inflammatory actions [90-94]. Beneficial effects of atorvastatin, mevastatin, rosuvastatin and simvastatin in acute ischemic injuries have been reported in human patients as well as in animal models [95-98]. Mechanism of statins’ actions include: (1) endothelial type III nitric oxide synthase (eNOS) up-regulation leads to inhibition of platelet activation; (2) reduce of malondialdehyde (MDA) and oxidized LDL (oxLDL); (3) alteration in the gene expression of inflammatory molecules such as VCAM-1, ICAM-1, E-selectin and interleukins [92, 93, 95]. As previously described, statins used for acute ischemic injuries with a different outcome. Some of these agents have better penetration into the cells, and it is because of the various lipophilic properties of this agents [99]. The use of statins has been reported with an increase in the risk of symptomatic hemorrhagic transformation (SHT) because of antithrombotic and fibrinolytic properties of these drugs [100, 101].

8.4. Melatonin
Melatonin or N-acetyl-5-methoxytryptamine is a neurohormone produced in the pineal gland that regulates circadian rhythm and has several clinical application[102]. Melatonin has low bioavalability (up to 56%) that is different from person to person. The plasma half-life is 20 to 50 minutes. Melatonin is metabolized by liver to at least 14 metabolites [103, 104]. Melatonin is the agonist of melatonin receptor 1 (MT1), melatonin receptor 2 (MT2) and nuclear receptor ROR-β [104, 105]. MT1 and MT2 are expressed in CNS and other body organs [105]. Expression of these receptors in the CNS leads to the regulation of central circadian rhythmicity [104]. Potential properties of melatonin like antioxidant effect, free radical scavenger, and anti-inflammatory reported in several studies [106-113]. Melatonin’s mechanisms of action as neuroprotective agents are as below:

1) alteration of antioxidant enzymes gene expressions like catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD) [114]. 2) attenuation of the activation of Nuclear Factor- Kappa B (NF-κB) and activator protein 1 (AP-1); downregulation of tumor necrosis factor alpha (TNFα), Cyclooxygenase2 (COX2), Interleukin 1β [102, 111]. 3) decrease in the level of phospho-Jun N-terminal Kinase 1 (p-JNK1) leads to suppression of apoptotic factors [102]. 4) direct detoxification of free radicals like hydroxyl and protection of the DNA by donation of the electron [115]. Melatonin showed good outcome as neuroprotective agents in various neuropathologies [106-113].

8.5. Erythropoietin
Erythropoietin is a cytokine and hormone that is produced by the kidneys and the liver. It can stimulate erythropoiesis [116] and maitains the blood hemoglobin concentration under different circumstances [117]. Erythropoietin has a bioavailability of 20-30% after subcutaneous administration. Plasma half-life of this drug is more than 24 hours. Elimination half-life is up to 13 hours after intravenous administration [118]. Erythropoietin is indicated for the treatment of anemia because of various etiologies such as chronic kidney disease, chemotherapy, blood loss and drug adverse events [117]. The role of erythropoietin as a neuroprotective agent is documented in some studies on animal models [119-123]. Expression of erythropoietin receptor (EpoR) in the brain tissue is responsible for the neuroprotective effect of this agent [121]. Neuroprotective action of erythropoietin occurs through three signaling pathways and leads to inhibition of apoptosis [124]. Erythropoietin readily crosses the blood-brain barrier (BBB) after brain insult and even through normal BBB by specific receptors [119, 123]. It activates (1) Janus tyrosine kinase 2 (JAK-2)-STAT signaling pathways that lead to the expression of Bcl-2 [125, 126], (2) extracellular-regulated kinase (ERK) and Protein kinase B (PKB), (3) nuclear factor-kappa B (NF-κB) [123, 124]. Recently, carbamylated erythropoietin has been reported as a neuroprotective agent that acts via the CD131/GDNF/AKT pathway in mice [126]. It does not bind to EPO-R and does not stimulate erythropoiesis nor activates JAK-2 pathways [127].

8.6. Free Radical Scavengers
Free radical scavengers such as polyethylene glycol (PEG)-conjugated SOD (PEG-SOD), 4-hydroxy- 2,2,6,6-tetramethylpiperidine-1-oxyl (Tempol), trans-2,3',4, 5'-tetrahydroxystilbene (hydroxystilbene oxyresveratrol) and disodium 2,4-disulfophenyl-N-tert-butylnitrone (NXY-059) have been used as neuroprotectants in various animal studies [128-133]. PEG-SOD, tempol, and hydroxystilbene oxyresveratrol did not show significant effects on neuropathologies [129, 130, 134]. NXY-059 as a scavenger of reactive oxygen species (ROS) has been used in rats and humans [128, 132, 133], and has been shown to exert antioxidant effects and vascular protective properties [128]. NXY-059 has been shown to be effective in the prevention of the salicylate oxidation [135]. Effectiveness of this drug is due to the entrapment of free radicals [135]. Around 80-90% of NXY-059 is eliminated unchanged through renal route. The elimination half-life is 2-4 hours in patients with normal kidney function [136].

8.7. Immunosuppressant drugs
Immunosuppressant drugs such as cyclosporine A (CsA) and particulaly tacrolimus (FK506) are recognized as neuroprotective agents in ischemic brain injuries and have been widely used in animal models [137-141]. Both cyclosporine and tacrolimus are calcineurin inhibitors. These agents bind to immunophilins and block the clacineurin that leads to reduced interleukin 2 production and T cells [142]. Tacrolimus is also a macrolid antibiotic and has more potency than cyclosporin with a different mode of immuphilin receptor inhibition [142]. Both drugs are substrate for cytochrome P450 3A4 and have potential renal and hepatic side effects [143]. One of the neuroprotective mechanisms of is blockage of extracellular signal-regulated kinases 1 and 2 (ERK1/2) [144]. ERK1 and 2 have pro-apoptotic properties and expression of these molecules occurs following the ischemic state [144]. FK506 inhibits calcineurin activity and nitric oxide (NO) production [140, 144]. Another mechanism of FK506 action is a reduction in the level of tumor necrosis factor-alpha (TNF-α) and IL-1beta [141] but it did not show anti-caspase-3 activity [145].

8.8. N-acetyl-L-cysteine (NAC)
NAC is an antidote for paracetamol toxicity and a thiol-containing drug with antioxidant, anti-inflammatory and free radical scavenging activity [146]. It is a safe medication with direct effects on glutathione synthesis. The main indication of NAC is in chronic bronchitis with hypersecretion of mucus, cystic fibrosis, acute respiratory distress syndrome and pulmonary oxygen toxicity. It can also attenuate brain oxidative stress. Infusion of NAC leads to the presence of the drug by up to 6 hours in plasma [146, 147]. Mechanisms of action of NAC as a neuroprotective agent are as below: 1) Increases the level of glutathione in the cells to prevent oxidative stress [146]. 2) Inhibition of Nitric oxide synthase (NOS) and increases tissue oxygenation [148, 149]. 3) Scavenging of the superoxide anions and ROS [149, 150]. 4) Inhibition of endothelial apoptosis and NF-κB, TNF-α activation [151, 152]. NAC can cross BBB, depending on the route of administration and the dosage of the drug [148]. The decrease of cerebral vasospasm is reported in a human patients and animal models with Subarachnoid hemorrhage after administration of NAC [148, 153, 154]. The beneficial effect of NAC has been reported in acute ischemic and hemorrhagic stroke and TBI in rodents [149, 152-155].

8.9. β-Blockers
Beta-blockers are a class of drugs that are widely used to reduce blood pressure and control cardiac arrhythmias through blockage of β-adrenergic receptors. [156]. Beta blockers are water- and fat-soluble. Water-soluble β-blockers have longer half lives and have renal elimination while fat-soluble ones have shorter half lives and are metabolized by the liver. This class of drugs have a good absorbtion via oral route. These drugs reduce cardiovascular morbidity and mortality and induce vasodilation through nitric oxide and receptor blockage [157]. The benefit of beta-blockers in TBI has been investigated in animal models and human patients [158-164]. Mechanisms of action of beta-blockers as neuroprotective agents are inhibition of apoptosis, attenuation of TNF-α and interleukin-1β expression and improvement the cortical microvascular perfusion [158].

8.10. COX-2 selective inhibitors
COX-2 selective inhibitors are blocking agents of the cyclooxygenase-2 enzyme, and are classified as a member of nonsteroidal anti-inflammatory drugs. COX-2 stimulates inflammation by converting arachidonic acid to prostaglandin [165] and activating NMDA receptors [166]. COX-2 has a significant excitotoxicity role through overproduction of prostaglandins[167]. All COX-2 inhibitors are metabolized by cytochrome P450 enzymes. They are used for the treatment of osteoarthritis, rheumatoid arthritis and painful conditions. These drugs have a lower risk of developing gastrointestinal side effects compared with non-selective COX inhibitors. COX-2 inhibitors have also protective activity against neurodegenerative diseases [168, 169]. Several animal studies have been performed on the effect of various COX-2 inhibitors such as valdecoxib, celecoxib, some natural products and NS-398 [170-172]. The use of COX-2 antagonists was associated with an increase in glutathione and superoxide dismutase levels, reduction in the levels of TNF-α, IL-1β and NF-κB [170], and blockage of NMDA receptors [167].

8.11. Curcumin
Curcumin is a natural polyphenolic compound with numerous medicinal properties [173]. Curcumin is a hydrophobic product with poor oral absorption and bioavailability. Commercial curcumin known as curcuminoids is composed of curcumin, demethoxycurcumin and bisdemethoxycurcumin [174]. Curcumin has two keto and enol tautomeric forms that affect the stability of the molecule [175]. Curcumin has anti-inflammatory [176-180], antioxidant [178, 181-185], immunomodulatory [186-189], anti-tumor and chemo-sensitizing [190-196], analgesic [197], lipid-modifying [198-202] and hepatoprotective [203-205] activities. Curcumin has been used for the treatment of TBI, ischemic and hemorrhagic stroke in animal models [206-209]. Various mechanisms have been suggested for the neuroprotective effects of curcumin. Zhu and colleagues reported that curcumin is an anti-inflammatory via “inhibition of Toll-like receptor 4 (TLR4) / Myeloid differentiation primary response gene 88 (MyD88) / NFκB signaling pathway” [206] in TBI. Curcumin can inhibit inflammatory processes in hemorrhagic strokes by a reduction in the expression of matrix metalloproteinases (MMPs), attenuation of IL-1 and inhibition of p38 mitogen-activated protein kinases (p38MAPK)/protein kinase C (PKC) pathways [207, 208]. Another mechanism of curcumin’s action in ischemic brain injuries is alteration of protein kinase B (Akt)/nuclear factorerythroid 2-related factor 2 (Nrf2) and reduction of oxidative damage [209]. Other neuroprotective agents such as corticosteroids [210, 211], barbiturates [212], ketamine [213], citicoline [214, 215], growth factors [216-219], minocycline [220, 221] and mannitol [222] with their mechanisms of action are summarized in (Table 2)

Table. Table 2
Table 2


  • Other potential neuroprotectants in the intensive care unit.
9. Conclusion
Neurological complications continue to be a major problem in patients admitted to ICU and significantly affect clinical outcomes as well as the length of ICU stay. Over the decades and centuries, numerous neuroprotective agents have been introduced to improve the care of critically ill patients. Despite the usefulness of these agents, none of them was really effective in the management of patients admitted to the ICU. The beneficial impact of various neuroprotective agents has been shown in animal models. Inhibition of damaging signaling pathways to the neurons such as inflammation, oxidative stress and apoptosis is the major molecular mechanism of neuroprotective agents. Use of neuroprotective agents in the ICU should be supported by compelling evidence on the improvement of clinical outcome and rapid recovery in the patients. However, the efficacy of agents discussed above is controversial in the light of findings of clinical trials. Some clinical trials have shown favorable clinical outcomes after the use of magnesium in patients with stroke. Melatonin and erythropoietin may be regarded as effective neuroprotective agents with anti-inflammatory and anti-apoptotic properties. Further studies in large populations of ICU patients should be performed to evaluate the neuroprotective effects of various agents such as curcumin, erythropoietin, magnesium and melatonin.

Conflict of interest
The authors declare that there are no conflicts of interest.
References
  1. Jain KK. The handbook of neuroprotection. Humana: New York; 2011.
  2. Porter D, Johnston AM, Henning J. Medical Conditions Requiring Intensive Care. Journal of the Royal Army Medical Corps. 2009;155:141-146.
  3. Peisker T, Koznar B, Stetkarova I, Widimsky P. Acute stroke therapy: A review. Trends in Cardiovascular Medicine. 2017;27:59-66.
  4. Tahir R, Pabaney A. Therapeutic hypothermia and ischemic stroke: A literature review. Surgical Neurology International. 2016;7:381.
  5. Gonzalez-Ibarra FP, Varon J, Lopez-Meza EG. Therapeutic hypothermia: critical review of the molecular mechanisms of action. Frontiers in Neurology. 2011;2:4.
  6. Great Britain. Department of Health. Comprehensive critical care: a review of adult critical care services. Department of Health: London 2000.
  7. Parrillo JE, Dellinger RP. Critical care medicine: principles of diagnosis and management in the adult. Elsevier Mosby: St. Louis, Mo.; London 2008.
  8. Chong J, Dumont T, Francis-Frank L, Balaan M. Sepsis and Septic Shock. Critical Care Nursing Quarterly. 2015;38:111-120.
  9. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, et al. Surviving Sepsis Campaign Guidelines Committee including The Pediatric S. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock, 2012. Intensive Care Medicine. 2013;39:165-228.
  10. Angus DC, van der Poll T. Severe Sepsis and Septic Shock. New England Journal of Medicine. 2013;369:840-851.
  11. Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, et al. International study of the prevalence and outcomes of infection in intensive care units. Journal of the American Medical Association. 2009;302:2323-9.
  12. Mock C, Lormand JD, Goosen J, Joshipura M, Peden M. Guidelines for essential trauma care. Geneva : World Health Organization; 2004.
  13. Shen Q, Hiebert JB, Hartwell J, Thimmesch AR, Pierce JD. Systematic Review of Traumatic Brain Injury and the Impact of Antioxidant Therapy on Clinical Outcomes. Worldviews on Evidence-Based Nursing. 2016;13:380-389.
  14. Gruenbaum SE, Zlotnik A, Gruenbaum BF, Hersey D, Bilotta F. Pharmacologic Neuroprotection for Functional Outcomes After Traumatic Brain Injury: A Systematic Review of the Clinical Literature. CNS Drugs. 2016;30:791-806.
  15. Park E, Bell JD, Baker AJ. Traumatic brain injury: Can the consequences be stopped? Canadian Medical Association Journal. 2008;178:1163-1170.
  16. Tromp G, Weinsheimer S, Ronkainen A, Kuivaniemi H. Molecular basis and genetic predisposition to intracranial aneurysm. Annals of Medicine. 2014;46:597-606.
  17. Wills S, Ronkainen A, van der Voet M, Kuivaniemi H, Helin K, Leinonen E, et al. Familial intracranial aneurysms: an analysis of 346 multiplex Finnish families. Stroke. 2003;34:1370-4.
  18. Guo Y, Li P, Guo Q, Shang K, Yan D, Du S, et al. Pathophysiology and Biomarkers in Acute Ischemic Stroke – A Review. Tropical Journal of Pharmaceutical Research. 2014;12:1097.
  19. Liou AKF, Clark RS, Henshall DC, Yin XM, Chen J. To die or not to die for neurons in ischemia, traumatic brain injury and epilepsy: a review on the stress-activated signaling pathways and apoptotic pathways. Progress in Neurobiology. 2003;69:103-142.
  20. Elmore S. Apoptosis: A Review of Programmed Cell Death. Toxicologic Pathology. 2007;35:495-516.
  21. Graham SH, Chen J. Programmed Cell Death in Cerebral Ischemia. Journal of Cerebral Blood Flow & Metabolism. 2001;99-109.
  22. Dong XX, Wang Y, Qin ZH. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacologica Sinica. 2009;30:379-387.
  23. Sims NR, Zaidan E. Biochemical changes associated with selective neuronal death following short-term cerebral ischaemia. International Journal of Biochemistry & Cell Biology. 1995;27:531-50.
  24. Ndountse LT, Chan HM. Role of N-methyl-D-aspartate receptors in polychlorinated biphenyl mediated neurotoxicity. Toxicology Letters. 2009;184:50-5.
  25. Wong PC, Cai H, Borchelt DR, Price DL. Genetically engineered mouse models of neurodegenerative diseases. Nature Neuroscience. 2002;5:633-9.
  26. Brown GC, Bal-Price A. Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Molecular Neurobiology. 2003;27:325-55.
  27. Parathath SR, Parathath S, Tsirka SE. Nitric oxide mediates neurodegeneration and breakdown of the blood-brain barrier in tPA-dependent excitotoxic injury in mice. Journal of Cell Science. 2006;119:339-49.
  28. Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D. Antioxidant therapy in acute central nervous system injury: current state. Pharmacological Reviews. 2002;54:271-84.
  29. Chen H, Yoshioka H, Kim GS, Jung JE, Okami N, Sakata H, et al. Oxidative Stress in Ischemic Brain Damage: Mechanisms of Cell Death and Potential Molecular Targets for Neuroprotection. Antioxidants & Redox Signaling. 2011;14:1505-1517.
  30. Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, et al. Antioxidant gene therapy against neuronal cell death. Pharmacology & Therapeutics. 2014;142:206-230.
  31. Huang J, Upadhyay UM, Tamargo RJ. Inflammation in stroke and focal cerebral ischemia. Surgical Neurology. 2006;66:232-245.
  32. Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular Mechanisms of Apoptosis in Cerebral Ischemia: Multiple Neuroprotective Opportunities. Molecular Neurobiology. 2007;37:7-38.
  33. Niizuma K, Endo H, Chan PH. Oxidative stress and mitochondrial dysfunction as determinants of ischemic neuronal death and survival. Journal of Neurochemistry. 2009;109:133-138.
  34. O'Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW. 1,026 Experimental treatments in acute stroke. Annals of Neurology. 2006;59:467-477.
  35. Danysz W, Parsons CG. Neuroprotective potential of ionotropic glutamate receptor antagonists. Neurotoxicity Research. 2002;4:119-26.
  36. Schauwecker PE. Neuroprotection by glutamate receptor antagonists against seizure-induced excitotoxic cell death in the aging brain. Experimental Neurology. 2010;224:207-218.
  37. Milani D, Cross JL, Anderton RS, Blacker DJ, Knuckey NW, Meloni BP. Neuroprotective efficacy of poly-arginine R18 and NA-1 (TAT-NR2B9c) peptides following transient middle cerebral artery occlusion in the rat. Neuroscience Research. 2017;114:9-15.
  38. Milani D, Knuckey NW, Anderton RS, Cross JL, Meloni BP. The R18 Polyarginine Peptide Is More Effective Than the TAT-NR2B9c (NA-1) Peptide When Administered 60 Minutes after Permanent Middle Cerebral Artery Occlusion in the Rat. Stroke Research and Treatment. 2016;2016:1-9.
  39. Cook DJ, Teves L, Tymianski M. A Translational Paradigm for the Preclinical Evaluation of the Stroke Neuroprotectant Tat-NR2B9c in Gyrencephalic Nonhuman Primates. Science Translational Medicine. 2012;4:154ra133-154ra133.
  40. Hill MD, Martin RH, Mikulis D, Wong JH, Silver FL, terBrugge KG, et al. Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): A phase 2, randomised, double-blind, placebo-controlled trial. The Lancet Neurology. 2012;11:942-950.
  41. Ayuso MI, Montaner J. Advanced neuroprotection for brain ischemia: an alternative approach to minimize stroke damage. Expert Opinion on Investigational Drugs. 2015;24:1137-1142.
  42. Marshall J, Wong KY, Rupasinghe CN, Tiwari R, Zhao X, Berberoglu ED, et al. Inhibition ofN-Methyl-d-aspartate-induced Retinal Neuronal Death by Polyarginine Peptides Is Linked to the Attenuation of Stress-induced Hyperpolarization of the Inner Mitochondrial Membrane Potential. Journal of Biological Chemistry. 2015;290:22030-22048.
  43. Meloni BP, Brookes LM, Clark VW, Cross JL, Edwards AB, Anderton RS, et al. Poly-Arginine and Arginine-Rich Peptides are Neuroprotective in Stroke Models. Journal of Cerebral Blood Flow & Metabolism. 2015;35:993-1004.
  44. Fugere M, Appel J, Houghten RA, Lindberg I, Day R. Short Polybasic Peptide Sequences Are Potent Inhibitors of PC5/6 and PC7: Use of Positional Scanning-Synthetic Peptide Combinatorial Libraries as a Tool for the Optimization of Inhibitory Sequences. Molecular Pharmacology. 2006;71:323-332.
  45. Akhtar MI, Ullah H, Hamid M. Magnesium, a drug of diverse use. Journal of Pakistan Medical Association. 2011;61:1220-5.
  46. Zhang X, Li Y, Del Gobbo LC, Rosanoff A, Wang J, Zhang W, Song Y. Effects of Magnesium Supplementation on Blood Pressure: A Meta-Analysis of Randomized Double-Blind Placebo-Controlled Trials. Hypertension. 2016;68:324-33.
  47. Simental-Mendia LE, Sahebkar A, Rodriguez-Moran M, Guerrero-Romero F. A systematic review and meta-analysis of randomized controlled trials on the effects of magnesium supplementation on insulin sensitivity and glucose control. Pharmacological Research. 2016;111:272-82.
  48. Sharma P, Chung C, Vizcaychipi M. Magnesium: The Neglected Electrolyte? A Clinical Review. Pharmacology & Pharmacy. 2014;05:762-772.
  49. McIntosh TK, Juhler M, Wieloch T. Novel pharmacologic strategies in the treatment of experimental traumatic brain injury: 1998. Journal of Neurotrauma. 1998;15:731-69.
  50. Memon ZI, Altura BT, Benjamin JL, Cracco RQ, Altura BM. Predictive value of serum ionized but not total magnesium levels in head injuries. Scandinavian Journal of Clinical and Laboratory Investigation. 1995;55:671-7.
  51. Afshari D, Moradian N, Rezaei M. Evaluation of the intravenous magnesium sulfate effect in clinical improvement of patients with acute ischemic stroke. Clinical Neurology and Neurosurgery. 2013;115:400-4.
  52. Bharosay A, Bharosay VV, Varma M, Saxena K, Sodani A, Saxena R. Correlation of Brain Biomarker Neuron Specific Enolase (NSE) with Degree of Disability and Neurological Worsening in Cerebrovascular Stroke. Indian J Clin Biochem. 2012;27:186-90.
  53. Gonzalez-Garcia S, Gonzalez-Quevedo A, Fernandez-Concepcion O, Pena-Sanchez M, Menendez-Sainz C, Hernandez-Diaz Z, et al. Short-term prognostic value of serum neuron specific enolase and S100B in acute stroke patients. Clinical Biochemistry, 2012;45:1302-7.
  54. Lee TM, Ivers NM, Bhatia S, Butt DA, Dorian P, et al. Improving stroke prevention therapy for patients with atrial fibrillation in primary care: protocol for a pragmatic, cluster-randomized trial. Implementation Science. 2016;11:159.
  55. Lip GYH. Optimizing stroke prevention in elderly patients with atrial fibrillation. Journal of Thrombosis and Haemostasis 2016;14:2121-2123.
  56. Mazurek M, Lip GY. To occlude or not? Left atrial appendage occlusion for stroke prevention in atrial fibrillation. Heart. 2017;103:93-95.
  57. Mizukoshi G, Katsura K-I, Katayama Y. Urinary 8-hydroxy-2′-deoxyguanosine and serum S100βin acute cardioembolic stroke patients. Neurological Research. 2013;27:644-646.
  58. Saver JL, Starkman S, Eckstein M, Stratton SJ, Pratt FD, Hamilton S, et al. Prehospital use of magnesium sulfate as neuroprotection in acute stroke. N Engl J Med. 2015;372:528-36.
  59. Singh H, Jalodia S, Gupta MS, Talapatra P, Gupta V, Singh I. Role of magnesium sulfate in neuroprotection in acute ischemic stroke. Ann Indian Acad Neurol. 2012;15:177-80.
  60. Talkachova A, Jaakkola J, Mustonen P, Kiviniemi T, Hartikainen JEK, Palomäki A, et al. Stroke as the First Manifestation of Atrial Fibrillation. Plos One. 2016;11:e0168010.
  61. Akdemir H, Kulakszoğlu EO, Tucer B, Menkü A, Postalc L, Günald Ö. Magnesium Sulfate Therapy for Cerebral Vasospasm After Aneurysmal Subarachnoid Hemorrhage. Neurosurgery Quarterly. 2009;19:35-39.
  62. Chen T, Carter BS. Role of magnesium sulfate in aneurysmal subarachnoid hemorrhage management: A meta-analysis of controlled clinical trials. Asian J Neurosurg. 2011;6:26-31.
  63. Hassan T, Nassar M, Elhadi SM, Radi WK. Effect of magnesium sulfate therapy on patients with aneurysmal subarachnoid hemorrhage using serum S100B protein as a prognostic marker. Neurosurg Rev. 2012;35:421-7;discussion 427.
  64. Muroi C, Terzic A, Fortunati M, Yonekawa Y, Keller E. Magnesium sulfate in the management of patients with aneurysmal subarachnoid hemorrhage: a randomized, placebo-controlled, dose-adapted trial. Surg Neurol. 2008;69:33-9;discussion 39.
  65. van den Bergh WM, Algra A, van Kooten F, Dirven CM, van Gijn J, Vermeulen M, et al. Magnesium sulfate in aneurysmal subarachnoid hemorrhage: a randomized controlled trial. Stroke. 2005;36:1011-5.
  66. Veyna RS, Seyfried D, Burke DG, Zimmerman C, Mlynarek M, Nichols V, et al. Magnesium sulfate therapy after aneurysmal subarachnoid hemorrhage. J Neurosurg. 2002;96:510-4.
  67. Westermaier T, Stetter C, Vince GH, Pham M, Tejon JP, Eriskat J, et al. Prophylactic intravenous magnesium sulfate for treatment of aneurysmal subarachnoid hemorrhage: a randomized, placebo-controlled, clinical study. Crit Care Med. 2010;38:1284-90.
  68. Wong GK, Poon WS, Chan MT, Boet R, Gin T, Ng SC, et al. Plasma magnesium concentrations and clinical outcomes in aneurysmal subarachnoid hemorrhage patients: post hoc analysis of intravenous magnesium sulphate for aneurysmal subarachnoid hemorrhage trial. Stroke. 2010;41:1841-4.
  69. Habgood MD, Bye N, Dziegielewska KM, Ek CJ, Lane MA, Potter A, et al. Changes in blood-brain barrier permeability to large and small molecules following traumatic brain injury in mice. Eur J Neurosci. 2007;25:231-8.
  70. Koch SM, Warters RD, Mehlhorn U. The simultaneous measurement of ionized and total calcium and ionized and total magnesium in intensive care unit patients. J Crit Care. 2002;17:203-5.
  71. Dabbagh OC, Aldawood AS, Arabi YM, Lone NA, Brits R, Pillay M. Magnesium supplementation and the potential association with mortality rates among critically ill non-cardiac patients. Saudi Med J. 2006;27:821-5.
  72. Mirrahimi B, Mortazavi A, Nouri M, Ketabchi E, Amirjamshidi A, Ashouri A, et al. Effect of magnesium on functional outcome and paraclinical parameters of patients undergoing supratentorial craniotomy for brain tumors: a randomized controlled trial. Acta Neurochir (Wien). 2015;157:985-91;discussion 991.
  73. James ML, Blessing R, Phillips-Bute BG, Bennett E, Laskowitz DT. S100B and brain natriuretic peptide predict functional neurological outcome after intracerebral haemorrhage. Biomarkers. 2009;14:388-94.
  74. Taylor F, Huffman MD, Macedo AF, Moore THM, Burke M, Davey Smith G, et al. Statins for the primary prevention of cardiovascular disease; 2013.
  75. Banach M, Serban C, Sahebkar A, Mikhailidis DP, Ursoniu S, Ray KK, et al. Impact of statin therapy on coronary plaque composition: a systematic review and meta-analysis of virtual histology intravascular ultrasound studies. BMC Med. 2015;13:229.
  76. Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, et al. Efficacy and safety of cholesterol-lowering treatment: Prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet, 2005;366:1267-78.
  77. Kavalipati N, Shah J, Ramakrishan A, Vasnawala H. Pleiotropic effects of statins. Indian J Endocrinol Metab. 2015;19:554-62.
  78. Bianconi V, Sahebkar A, Banach M, Pirro M. Statins, haemostatic factors and thrombotic risk. Curr Opin Cardiol; 2017.
  79. Chrusciel P, Sahebkar A, Rembek-Wieliczko M, Serban MC, Ursoniu S, Mikhailidis DP, et al. Impact of statin therapy on plasma adiponectin concentrations: A systematic review and meta-analysis of 43 randomized controlled trial arms. Atherosclerosis. 2016;253:194-208.
  80. Derosa G, Maffioli P, Reiner Z, Simental-Mendia LE, Sahebkar A. Impact of Statin Therapy on Plasma Uric Acid Concentrations: A Systematic Review and Meta-Analysis. Drugs. 2016;76:947-56.
  81. Sahebkar A, Rathouska J, Derosa G, Maffioli P, Nachtigal P. Statin impact on disease activity and C-reactive protein concentrations in systemic lupus erythematosus patients: A systematic review and meta-analysis of controlled trials. Autoimmun Rev. 2016;15:344-53.
  82. Sahebkar A, Serban C, Ursoniu S, Mikhailidis DP, Undas A, Lip GY, et al. The impact of statin therapy on plasma levels of von Willebrand factor antigen. Systematic review and meta-analysis of randomised placebo-controlled trials. Thromb Haemost. 2016;115:520-32.
  83. Sahebkar A, Rathouska J, Simental-Mendia LE, Nachtigal P. Statin therapy and plasma cortisol concentrations: A systematic review and meta-analysis of randomized placebo-controlled trials. Pharmacol Res. 2016;103:17-25.
  84. Ferretti G, Bacchetti T, Sahebkar A. Effect of statin therapy on paraoxonase-1 status: A systematic review and meta-analysis of 25 clinical trials. Prog Lipid Res. 2015;60:50-73.
  85. Sahebkar A, Ponziani MC, Goitre I, Bo S. Does statin therapy reduce plasma VEGF levels in humans? A systematic review and meta-analysis of randomized controlled trials. Metabolism. 2015;64:1466-76.
  86. Sahebkar A, Kotani K, Serban C, Ursoniu S, Mikhailidis DP, Jones SR, et al. Statin therapy reduces plasma endothelin-1 concentrations: A meta-analysis of 15 randomized controlled trials. Atherosclerosis. 2015;241:433-42.
  87. Serban C, Sahebkar A, Ursoniu S, Mikhailidis DP, Rizzo M, Lip GY, et al. A systematic review and meta-analysis of the effect of statins on plasma asymmetric dimethylarginine concentrations. Sci Rep. 2015;5:9902.
  88. Sahebkar A, Serban C, Mikhailidis DP, Undas A, Lip GYH, Muntner P, et al. Association between statin use and plasma d-dimer levels: A systematic review and meta-analysis of randomised controlled trials. Thrombosis and Haemostasis. 2015;114:546-557.
  89. Sirtori CR. The pharmacology of statins. Pharmacol Res. 2014;88:3-11.
  90. Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ, et al. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat Med. 2000;6:1004-10.
  91. Asahi M, Huang Z, Thomas S, Yoshimura S, Sumii T, Mori T, et al. Protective effects of statins involving both eNOS and tPA in focal cerebral ischemia. J Cereb Blood Flow Metab. 2005;25:722-9.
  92. Jain MK, Ridker PM. Anti-Inflammatory Effects of Statins: Clinical Evidence and Basic Mechanisms. Nature Reviews Drug Discovery. 2005;4:977-987.
  93. Moon GJ, Kim SJ, Cho YH, Ryoo S, Bang OY. Antioxidant effects of statins in patients with atherosclerotic cerebrovascular disease. J Clin Neurol. 2014;10:140-7.
  94. Parizadeh SMR, Azarpazhooh MR, Moohebati M, Nematy M, Ghayour-Mobarhan M, Tavallaie S, et al. Simvastatin therapy reduces prooxidant-antioxidant balance: Results of a placebo-controlled cross-over trial. Lipids. 2011;46:333-340.
  95. Laufs U, Gertz K, Huang P, Nickenig G, Bohm M, Dirnagl U, et al. Atorvastatin upregulates type III nitric oxide synthase in thrombocytes, decreases platelet activation, and protects from cerebral ischemia in normocholesterolemic mice. Stroke. 2000;31:2442-9.
  96. Amin-Hanjani S, Stagliano NE, Yamada M, Huang PL, Liao JK, Moskowitz MA. Mevastatin, an HMG-CoA reductase inhibitor, reduces stroke damage and upregulates endothelial nitric oxide synthase in mice. Stroke. 2001;32:980-6.
  97. Moon GJ, Kim SJ, Cho YH, Ryoo S, Bang OY. Antioxidant Effects of Statins in Patients with Atherosclerotic Cerebrovascular Disease. Journal of Clinical Neurology. 2014;10:140.
  98. Montaner J, Chacón P, Krupinski J, Rubio F, Millán M, Molina CA, et al. Simvastatin in the acute phase of ischemic stroke: a safety and efficacy pilot trial. European Journal of Neurology. 2007;15:82-90.
  99. Endres M, Laufs U, Huang Z, Nakamura T, Huang P, Moskowitz MA, et al. Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 1998;95:8880-5.
  100. Cordenier A, De Smedt A, Brouns R, Uyttenboogaart M, De Raedt S, Luijckx GJ, et al. Pre-stroke use of statins on stroke outcome: a meta-analysis of observational studies. Acta Neurol Belg. 2011;111:261-7.
  101. Hong KS, Lee JS. Statins in Acute Ischemic Stroke: A Systematic Review. Journal of Stroke. 2015;17:282-301.
  102. Ali T, Badshah H, Kim TH, Kim MO. Melatonin attenuates D-galactose-induced memory impairment, neuroinflammation and neurodegeneration via RAGE/NF-KB/JNK signaling pathway in aging mouse model. Journal of Pineal Research. 2015;58:71-85.
  103. Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N, et al. Melatonin: Pharmacology, Functions and Therapeutic Benefits. Current Neuropharmacology. 2017;15:434-443.
  104. Rios ER, Venancio ET, Rocha NF, Woods DJ, Vasconcelos S, Macedo D, et al. Melatonin: pharmacological aspects and clinical trends. Int J Neurosci. 2010;120:583-90.
  105. Boutin JA, Audinot V, Ferry G, Delagrange P. Molecular tools to study melatonin pathways and actions. Trends Pharmacol Sci. 2005;26:412-9.
  106. Watson N, Diamandis T, Gonzales-Portillo C, Reyes S, Borlongan CV. Melatonin as an Antioxidant for Stroke Neuroprotection. Cell Transplantation. 2016;25:883-891.
  107. Bandyopadhyay D, Biswas K, Bandyopadhyay U, Reiter RJ, Banerjee RK. Melatonin protects against stress-induced gastric lesions by scavenging the hydroxyl radical. J Pineal Res. 2000;29:143-51.
  108. Chahbouni M, Escames G, Venegas C, Sevilla B, García JA, López LC, et al. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. Journal of Pineal Research. 2010;48:282-289.
  109. Pei Z, Fung PC, Cheung RT. Melatonin reduces nitric oxide level during ischemia but not blood-brain barrier breakdown during reperfusion in a rat middle cerebral artery occlusion stroke model. J Pineal Res. 2003;34:110-8.
  110. Koh PO. Melatonin regulates nitric oxide synthase expression in ischemic brain injury. J Vet Med Sci. 2008;70:747-50.
  111. Beni SM. Melatonin-induced neuroprotection after closed head injury is associated with increased brain antioxidants and attenuated late-phase activation of NF- B and AP-1. The FASEB Journal; 2003.
  112. Ozdemir D, Uysal N, Gonenc S, Acikgoz O, Sonmez A, Topcu A, et al. Effect of melatonin on brain oxidative damage induced by traumatic brain injury in immature rats. Physiol Res. 2005;54:631-7.
  113. Ozdemir D, Tugyan K, Uysal N, Sonmez U, Sonmez A, Acikgoz O, et al. Protective effect of melatonin against head trauma-induced hippocampal damage and spatial memory deficits in immature rats. Neuroscience Letters. 2005;385:234-239.
  114. Fischer TW, Kleszczyński K, Hardkop LH, Kruse N, Zillikens D. Melatonin enhances antioxidative enzyme gene expression (CAT, GPx, SOD), prevents their UVR-induced depletion, and protects against the formation of DNA damage (8-hydroxy-2'-deoxyguanosine) in ex vivo human skin. Journal of Pineal Research. 2013;54:303-312.
  115. Reiter RJ, Mayo JC, Tan D-X, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. Journal of Pineal Research. 2016;61:253-278.
  116. Chung E, Kong X, Goldberg MP, Stowe AM, Raman L. Erythropoietin-mediated neuroprotection in a pediatric mouse model of chronic hypoxia. Neurosci Lett. 2015;597:54-9.
  117. Jelkmann W. Physiology and Pharmacology of Erythropoietin. Transfusion Medicine and Hemotherapy. 2013;40:302-309.
  118. Jurado Garcia JM, Torres Sanchez E, Olmos Hidalgo D, Alba Conejo E. Erythropoietin pharmacology. Clin Transl Oncol. 2007;9:715-22.
  119. Grasso G, Buemi M, Alafaci C, Sfacteria A, Passalacqua M, Sturiale A, et al. Beneficial effects of systemic administration of recombinant human erythropoietin in rabbits subjected to subarachnoid hemorrhage. Proc Natl Acad Sci U S A. 2002;99:5627-31.
  120. Chen G, Zhang S, Shi J, Ai J, Hang C. Effects of recombinant human erythropoietin (rhEPO) on JAK2/STAT3 pathway and endothelial apoptosis in the rabbit basilar artery after subarachnoid hemorrhage. Cytokine. 2009;45:162-168.
  121. Sanchez PE, Fares RP, Risso JJ, Bonnet C, Bouvard S, Le-Cavorsin M, et al. Optimal neuroprotection by erythropoietin requires elevated expression of its receptor in neurons. Proc Natl Acad Sci U S A. 2009;106:9848-53.
  122. Taoufik E, Petit E, Divoux D, Tseveleki V, Mengozzi M, Roberts ML, et al. TNF receptor I sensitizes neurons to erythropoietin- and VEGF-mediated neuroprotection after ischemic and excitotoxic injury. Proceedings of the National Academy of Sciences. 2008;105:6185-6190.
  123. Brines ML, Ghezzi P, Keenan S, Agnello D, de Lanerolle NC, Cerami C, et al. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proceedings of the National Academy of Sciences. 2000;97:10526-10531.
  124. Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci. 2005;6:484-94.
  125. Sepúlveda P, Encabo A, Carbonell-Uberos F, Miñana MD. BCL-2 expression is mainly regulated by JAK/STAT3 pathway in human CD34+ hematopoietic cells. Cell Death and Differentiation. 2006;14:378-380.
  126. Ding J, Wang J, Li QY, Yu JZ, Ma CG, Wang X, et al. Neuroprotection and CD131/GDNF/AKT Pathway of Carbamylated Erythropoietin in Hypoxic Neurons. Mol Neurobiol; 2016.
  127. Chen J, Chen J, Yang Z, Zhang X. Carbamylated Erythropoietin: A Prospective Drug Candidate for Neuroprotection. Biochemistry Insights. 2016;25.
  128. Clausen F, Marklund N, Lewen A, Hillered L. The nitrone free radical scavenger NXY-059 is neuroprotective when administered after traumatic brain injury in the rat. J Neurotrauma. 2008;25:1449-57.
  129. Kwon TH, Chao DL, Malloy K, Sun D, Alessandri B, Bullock MR. Tempol, a novel stable nitroxide, reduces brain damage and free radical production, after acute subdural hematoma in the rat. J Neurotrauma. 2003;20:337-45.
  130. Kato N, Yanaka K, Hyodo K, Homma K, Nagase S, Nose T. Stable nitroxide Tempol ameliorates brain injury by inhibiting lipid peroxidation in a rat model of transient focal cerebral ischemia. Brain Res. 2003;979:188-93.
  131. Rak R, Chao DL, Pluta RM, Mitchell JB, Oldfield EH, Watson JC. Neuroprotection by the stable nitroxide Tempol during reperfusion in a rat model of transient focal ischemia. J Neurosurg. 2000;92:646-51.
  132. Lees KR, Zivin JA, Ashwood T, Davalos A, Davis SM, Diener H-C, et al. NXY-059 for Acute Ischemic Stroke. New England Journal of Medicine. 2006;354:588-600.
  133. Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, et al. NXY-059 for the Treatment of Acute Ischemic Stroke. New England Journal of Medicine. 2007;357:562-571.
  134. Lorenz P, Roychowdhury S, Engelmann M, Wolf G, Horn TF. Oxyresveratrol and resveratrol are potent antioxidants and free radical scavengers: effect on nitrosative and oxidative stress derived from microglial cells. Nitric Oxide. 2003;9:64-76.
  135. Maples KR, Ma F, Zhang YK. Comparison of the radical trapping ability of PBN, S-PPBN and NXY-059. Free Radic Res. 2001;34:417-26.
  136. Strid S, Borga O, Edenius C, Jostell KG, Odergren T, Weil A. Pharmacokinetics in renally impaired subjects of NXY-059, a nitrone-based, free-radical trapping agent developed for the treatment of acute stroke. Eur J Clin Pharmacol. 2002;58:409-15.
  137. Uchino H, Minamikawa-Tachino R, Kristian T, Perkins G, Narazaki M, Siesjo BK, et al. Differential neuroprotection by cyclosporin A and FK506 following ischemia corresponds with differing abilities to inhibit calcineurin and the mitochondrial permeability transition. Neurobiol Dis. 2002;10:219-33.
  138. Arii T, Kamiya T, Arii K, Ueda M, Nito C, Katsura K-I, et al. Neuroprotective effect of immunosuppressant FK506 in transient focal ischemia in rats: Therapeutic time window for FK506 in transient focal ischemia. Neurological Research. 2013;23:755-760.
  139. Saganová K, Gálik J, Blaško J, Korimová A, Račeková E, Vanický I. Immunosuppressant FK506: Focusing on neuroprotective effects following brain and spinal cord injury. Life Sciences. 2012;91:77-82.
  140. Sharifi Z-N, Abolhassani F, Zarrindast MR, Movassaghi S, Rahimian N, Hassanzadeh G. Effects of FK506 on Hippocampal CA1 Cells Following Transient Global Ischemia/Reperfusion in Wistar Rat. Stroke Research and Treatment. 2012;2012:1-8.
  141. Zawadzka M, Kaminska B. A novel mechanism of FK506-mediated neuroprotection: Downregulation of cytokine expression in glial cells. Glia. 2005;49:36-51.
  142. Pillans P. Experimental and Clinical Pharmacology: Immunosuppressants - mechanisms of action and monitoring. Australian Prescriber. 2006;29:99-101.
  143. Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med. 2004;351:2715-29.
  144. Szydlowska K, Gozdz A, Dabrowski M, Zawadzka M, Kaminska B. Prolonged activation of ERK triggers glutamate-induced apoptosis of astrocytes: neuroprotective effect of FK506. Journal of Neurochemistry. 2010;113:904-918.
  145. Muramoto M, Yamazaki T, Nishimura S, Kita Y. Detailed in vitro pharmacological analysis of FK506-induced neuroprotection. Neuropharmacology. 2003;45:394-403.
  146. Arakawa M, Ito Y. N-acetylcysteine and neurodegenerative diseases: Basic and clinical pharmacology. The Cerebellum. 2007;6:308-314.
  147. Elbini Dhouib I, Jallouli M, Annabi A, Gharbi N, Elfazaa S, Lasram MM. A minireview on N-acetylcysteine: An old drug with new approaches. Life Sciences. 2016;151:359-363.
  148. Bavarsad Shahripour R, Harrigan MR, Alexandrov AV. N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities. Brain and Behavior. 2014;4:108-122.
  149. Cuzzocrea S, Mazzon E, Costantino G, Serraino I, Dugo L, Calabrò G, et al. Beneficial effects ofn-acetylcysteine on ischaemic brain injury. British Journal of Pharmacology. 2000;130:1219-1226.
  150. Sen O, Caner H, Aydin MV, Ozen O, Atalay B, Altinors N, et al. The effect of mexiletine on the level of lipid peroxidation and apoptosis of endothelium following experimental subarachnoid hemorrhage. Neurol Res. 2006;28:859-63.
  151. Findlay JM, Weir BK, Kanamaru K, Espinosa F. Arterial wall changes in cerebral vasospasm. Neurosurgery. 1989;25:736-45; discussion 745-6.
  152. Chen G, Shi J, Hu Z, Hang C. Inhibitory effect on cerebral inflammatory response following traumatic brain injury in rats: a potential neuroprotective mechanism of N-acetylcysteine. Mediators Inflamm. 2008;2008:716458.
  153. Guney O, Erdi F, Esen H, Kiyici A, Kocaogullar Y. N-acetylcysteine prevents vasospasm after subarachnoid hemorrhage. World Neurosurg. 2010;73:42-9;discussion e3.
  154. Pereira Filho Nde A, Pereira Filho Ade A, Soares FP, Coutinho LM. Effect of N-acetylcysteine on vasospasm in subarachnoid hemorrhage. Arq Neuropsiquiatr. 2010;68:918-22.
  155. Akca T, Canbaz H, Tataroglu C, Caglikulekci M, Tamer L, Colak T, et al. The Effect of N-Acetylcysteine on Pulmonary Lipid Peroxidation and Tissue Damage. Journal of Surgical Research. 2005;129:38-45.
  156. Frishman WH, Saunders E. beta-Adrenergic blockers. J Clin Hypertens (Greenwich). 2011;13:649-53.
  157. Koch-Weser J, Frishman WH. beta-Adrenoceptor antagonists: new drugs and new indications. N Engl J Med. 1981;305:500-6.
  158. Savitz SI, Erhardt JA, Anthony JV, Gupta G, Li X, Barone FC, et al. The novel beta-blocker, carvedilol, provides neuroprotection in transient focal stroke. J Cereb Blood Flow Metab. 2000;20:1197-204.
  159. Schroeppel TJ, Fischer PE, Zarzaur BL, Magnotti LJ, Clement LP, Fabian TC, et al. Beta-adrenergic blockade and traumatic brain injury: protective? J Trauma. 2010;69:776-82.
  160. Salim A, Hadjizacharia P, Brown C, Inaba K, Teixeira PGR, Chan L, et al. Significance of Troponin Elevation After Severe Traumatic Brain Injury. The Journal of Trauma: Injury, Infection, and Critical Care. 2008;64:46-52.
  161. Riordan WP, Cotton BA, Norris PR, Waitman LR, Jenkins JM, Morris JA. β-Blocker Exposure in Patients With Severe Traumatic Brain Injury (TBI) and Cardiac Uncoupling. The Journal of Trauma: Injury, Infection, and Critical Care. 2007;63:503-511.
  162. Inaba K, Teixeira PGR, David J-S, Chan LS, Salim A, Brown C, et al. Beta-Blockers in Isolated Blunt Head Injury. Journal of the American College of Surgeons. 2008;206:432-438.
  163. Hadjizacharia P, O'Keeffe T, Brown CV, Inaba K, Salim A, Chan LS, et al. Incidence, risk factors, and outcomes for atrial arrhythmias in trauma patients. Am Surg. 2011;77:634-9.
  164. Cotton BA, Snodgrass KB, Fleming SB, Carpenter RO, Kemp CD, Arbogast PG, et al. Beta-Blocker Exposure is Associated With Improved Survival After Severe Traumatic Brain Injury. The Journal of Trauma: Injury, Infection, and Critical Care. 2007;62:26-35.
  165. Chakraborti AK, Garg SK, Kumar R, Motiwala HF, Jadhavar PS. Progress in COX-2 inhibitors: a journey so far. Curr Med Chem. 2010;17:1563-93.
  166. Manabe Y, Anrather J, Kawano T, Niwa K, Zhou P, Ross ME, et al. Prostanoids, not reactive oxygen species, mediate COX-2-dependent neurotoxicity. Annals of Neurology. 2004;55:668-675.
  167. Stark DT, Bazan NG. Synaptic and extrasynaptic NMDA receptors differentially modulate neuronal cyclooxygenase-2 function, lipid peroxidation, and neuroprotection. J Neurosci. 2011;31:13710-21.
  168. Capone ML, Tacconelli S, Sciulli MG, Patrignani P. Clinical pharmacology of selective COX-2 inhibitors. Int J Immunopathol Pharmacol. 2003;16:49-58.
  169. Bertolini A, Ottani A, Sandrini M. Selective COX-2 inhibitors and dual acting anti-inflammatory drugs: critical remarks. Curr Med Chem. 2002;9:1033-43.
  170. Singh DP, Chopra K. Flavocoxid, dual inhibitor of cyclooxygenase-2 and 5-lipoxygenase, exhibits neuroprotection in rat model of ischaemic stroke. Pharmacol Biochem Behav. 2014;120:33-42.
  171. Ahmad M, Zhang Y, Liu H, Rose ME, Graham SH. Prolonged opportunity for neuroprotection in experimental stroke with selective blockade of cyclooxygenase-2 activity. Brain Research. 2009;1279:168-173.
  172. Vinukonda G, Csiszar A, Hu F, Dummula K, Pandey NK, Zia MT, et al. Neuroprotection in a rabbit model of intraventricular haemorrhage by cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-alpha inhibition. Brain. 2010;133:2264-2280.
  173. Sahebkar A. Curcumin: A Natural Multitarget Treatment for Pancreatic Cancer. Integrative Cancer Therapies. 2016;15:333-334.
  174. Huang S, Beevers CS. Pharmacological and clinical properties of curcumin. Botanics: Targets and Therapy. 2011;5.
  175. Mullaicharam AR, Maheswaran A. Pharmacological effects of curcumin. International journal of Nutrition, Pharmacology, Neurological Diseases. 2012;2:92.
  176. Ghandadi M, Sahebkar A. Curcumin: An effective inhibitor of interleukin-6. Curr Pharm Des; 2016.
  177. Panahi Y, Hosseini MS, Khalili N, Naimi E, Simental-Mendia LE, Majeed M, et al. Effects of curcumin on serum cytokine concentrations in subjects with metabolic syndrome: A post-hoc analysis of a randomized controlled trial. Biomed Pharmacother. 2016;82:578-82.
  178. Panahi Y, Hosseini MS, Khalili N, Naimi E, Majeed M, Sahebkar A. Antioxidant and anti-inflammatory effects of curcuminoid-piperine combination in subjects with metabolic syndrome: A randomized controlled trial and an updated meta-analysis. Clin Nutr. 2015;34:1101-8.
  179. Sahebkar A. Are curcuminoids effective C-reactive protein-lowering agents in clinical practice? Evidence from a meta-analysis. Phytother Res. 2014;28:633-42.
  180. Panahi Y, Sahebkar A, Parvin S, Saadat A. A randomized controlled trial on the anti-inflammatory effects of curcumin in patients with chronic sulphur mustard-induced cutaneous complications. Ann Clin Biochem. 2012;49:580-8.
  181. Panahi Y, Khalili N, Sahebi E, Namazi S, Karimian MS, Majeed M, et al. Antioxidant effects of curcuminoids in patients with type 2 diabetes mellitus: a randomized controlled trial. Inflammopharmacology. 2017;25:25-31.
  182. Panahi Y, Alishiri GH, Parvin S, Sahebkar A. Mitigation of Systemic Oxidative Stress by Curcuminoids in Osteoarthritis: Results of a Randomized Controlled Trial. J Diet Suppl. 2016;13:209-20.
  183. Panahi Y, Ghanei M, Hajhashemi A, Sahebkar A. Effects of Curcuminoids-Piperine Combination on Systemic Oxidative Stress, Clinical Symptoms and Quality of Life in Subjects with Chronic Pulmonary Complications Due to Sulfur Mustard: A Randomized Controlled Trial. J Diet Suppl. 2016;13:93-105.
  184. Sahebkar A, Mohammadi A, Atabati A, Rahiman S, Tavallaie S, Iranshahi M, et al. Curcuminoids modulate pro-oxidant-antioxidant balance but not the immune response to heat shock protein 27 and oxidized LDL in obese individuals. Phytother Res. 2013;27:1883-8.
  185. Panahi Y, Sahebkar A, Amiri M, Davoudi SM, Beiraghdar F, Hoseininejad SL, et al. Improvement of sulphur mustard-induced chronic pruritus, quality of life and antioxidant status by curcumin: results of a randomised, double-blind, placebo-controlled trial. Br J Nutr. 2012;108:1272-9.
  186. Abdollahi E, Momtazi AA, Johnston TP, Sahebkar A. Therapeutic Effects of Curcumin in Inflammatory and Immune-Mediated Diseases: A Nature-Made Jack-of-All-Trades? J Cell Physiol; 2017.
  187. Karimian MS, Pirro M, Majeed M, Sahebkar A. Curcumin as a natural regulator of monocyte chemoattractant protein-1. Cytokine Growth Factor Rev; 2016.
  188. Derosa G, Maffioli P, Simental-Mendia LE, Bo S, Sahebkar A. Effect of curcumin on circulating interleukin-6 concentrations: A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2016;111:394-404.
  189. Sahebkar A, Cicero AF, Simental-Mendia LE, Aggarwal BB, Gupta SC. Curcumin downregulates human tumor necrosis factor-alpha levels: A systematic review and meta-analysis ofrandomized controlled trials. Pharmacol Res. 2016;107:234-42.
  190. Lelli D, Pedone C, Sahebkar A. Curcumin and treatment of melanoma: The potential role of microRNAs. Biomed Pharmacother. 2017;88:832-834.
  191. Ramezani M, Hatamipour M, Sahebkar A. Promising Anti-tumor properties of Bisdemethoxycurcumin: A Naturally Occurring Curcumin Analogue. J Cell Physiol; 2017.
  192. Momtazi AA, Shahabipour F, Khatibi S, Johnston TP, Pirro M, Sahebkar A. Curcumin as a MicroRNA Regulator in Cancer: A Review. Rev Physiol Biochem Pharmacol. 2016;171:1-38.
  193. Momtazi AA, Sahebkar A. Difluorinated Curcumin: A Promising Curcumin Analogue with Improved Anti-Tumor Activity and Pharmacokinetic Profile. Curr Pharm Des. 2016;22:4386-97.
  194. Rezaee R, Momtazi AA, Monemi A, Sahebkar A. Curcumin: A potentially powerful tool to reverse cisplatin-induced toxicity. Pharmacol Res. 2017;117:218-227.
  195. Teymouri M, Pirro M, Johnston TP, Sahebkar A. Curcumin as a multifaceted compound against human papilloma virus infection and cervical cancers: A review of chemistry, cellular, molecular, and preclinical features. Biofactors; 2016.
  196. Mirzaei H, Naseri G, Rezaee R, Mohammadi M, Banikazemi Z, Mirzaei HR, et al. Curcumin: A new candidate for melanoma therapy? Int J Cancer. 2016;139:1683-95.
  197. Sahebkar A, Henrotin Y. Analgesic efficacy and safety of curcuminoids in clinical practice: A systematic review and meta-analysis of randomized controlled trials. Pain Medicine (United States). 2016;17:1192-1202.
  198. Panahi Y, Kianpour P, Mohtashami R, Jafari R, Simental-Mendia LE, Sahebkar A. Curcumin Lowers Serum Lipids and Uric Acid in Subjects With Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial. J Cardiovasc Pharmacol. 2016;68:223-9.
  199. Ganjali S, Blesso CN, Banach M, Pirro M, Majeed M, Sahebkar A. Effects of curcumin on HDL functionality. Pharmacol Res. 2017;119:208-218.
  200. Panahi Y, Khalili N, Hosseini MS, Abbasinazari M, Sahebkar A. Lipid-modifying effects of adjunctive therapy with curcuminoids-piperine combination in patients with metabolic syndrome: results of a randomized controlled trial. Complement Ther Med. 2014;22:851-7.
  201. Sahebkar A. Curcuminoids for the management of hypertriglyceridaemia. Nat Rev Cardiol. 2014;11:123.
  202. Cicero AFG, Colletti A, Bajraktari G, Descamps O, Djuric DM, Ezhov M, et al. Lipid lowering nutraceuticals in clinical practice: Position paper from an International Lipid Expert Panel. Archives of Medical Science. 2017;13:965-1005.
  203. Zabihi NA, Pirro M, Johnston TP, Sahebkar A. Is there a role for curcumin supplementation in the treatment of non-alcoholic fatty liver disease? The data suggest yes. Curr Pharm Des; 2016.
  204. Rahmani S, Asgary S, Askari G, Keshvari M, Hatamipour M, Feizi A, et al. Treatment of Non-alcoholic Fatty Liver Disease with Curcumin: A Randomized Placebo-controlled Trial. Phytother Res. 2016;30:1540-8.
  205. Panahi Y, Kianpour P, Mohtashami R, Jafari R, Simental-Mendia LE, Sahebkar A. Efficacy and Safety of Phytosomal Curcumin in Non-Alcoholic Fatty Liver Disease: A Randomized Controlled Trial. Drug Res (Stuttg); 2017.
  206. Zhu HT, Bian C, Yuan JC, Chu WH, Xiang X, Chen F, et al. Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-kappaB signaling pathway in experimental traumatic brain injury. J Neuroinflammation. 2014;11:59.
  207. Sun Y, Dai M, Wang Y, Wang W, Sun Q, Yang G-Y, et al. Neuroprotection and Sensorimotor Functional Improvement by Curcumin after Intracerebral Hemorrhage in Mice. Journal of Neurotrauma. 2011;28:2513-2521.
  208. Yang Z, Zhao T, Zou Y, Zhang JH, Feng H. Curcumin inhibits microglia inflammation and confers neuroprotection in intracerebral hemorrhage. Immunology Letters. 2014;160:89-95.
  209. Arai K, Wu J, Li Q, Wang X, Yu S, Li L, et al. Neuroprotection by Curcumin in Ischemic Brain Injury Involves the Akt/Nrf2 Pathway. PLoS ONE. 2013;8:e59843.
  210. Alderson P, Roberts I. Corticosteroids in acute traumatic brain injury: systematic review of randomised controlled trials. BMJ. 1997;314:1855-9.
  211. Sandercock PAG, Soane T, Sandercock PAG. Corticosteroids for acute ischaemic stroke; 2011.
  212. Roberts I, Sydenham E, Roberts I. Barbiturates for acute traumatic brain injury; 2012.
  213. Bell JD. In Vogue: Ketamine for Neuroprotection in Acute Neurologic Injury. Anesthesia & Analgesia. 2017;1.
  214. Adibhatla RM, Hatcher JF. Citicoline mechanisms and clinical efficacy in cerebral ischemia. J Neurosci Res. 2002;70:133-9.
  215. Hurtado O, Hernandez-Jimenez M, Zarruk JG, Cuartero MI, Ballesteros I, Camarero G, et al. Citicoline (CDP-choline) increases Sirtuin1 expression concomitant to neuroprotection in experimental stroke. J Neurochem. 2013;126:819-26.
  216. Subirós N, Pérez-Saad H, Aldana L, Gibson CL, Borgnakke WS, Garcia-del-Barco D. Neuroprotective effect of epidermal growth factor plus growth hormone-releasing peptide-6 resembles hypothermia in experimental stroke. Neurological Research. 2016;38:950-958.
  217. Sofroniew MV, Howe CL, Mobley WC. Nerve Growth Factor Signaling, Neuroprotection, and Neural Repair. Annual Review of Neuroscience. 2001;24:1217-1281.
  218. Alzheimer C, Werner S. Fibroblast growth factors and neuroprotection. Adv Exp Med Biol. 2002;513:335-51.
  219. Gora-Kupilas K, Josko J. The neuroprotective function of vascular endothelial growth factor (VEGF). Folia Neuropathol. 2005;43:31-9.
  220. Plane JM, Shen Y, Pleasure DE, Deng W. Prospects for Minocycline Neuroprotection. Archives of Neurology. 2010;67.
  221. Amiri-Nikpour MR, Nazarbaghi S, Hamdi-Holasou M, Rezaei Y. An open-label evaluator-blinded clinical study of minocycline neuroprotection in ischemic stroke: gender-dependent effect. Acta Neurologica Scandinavica. 2015;131:45-50.
  222. Wakai A, McCabe A, Roberts I, Schierhout G, Wakai A. Mannitol for acute traumatic brain injury; 2013.
  223. Aydin MV, Caner H, Sen O, Ozen O, Atalay B, Cekinmez M, et al. Effect of melatonin on cerebral vasospasm following experimental subarachnoid hemorrhage. Neurological Research. 2013;27:77-82.
  224. Ayer RE, Sugawara T, Chen W, Tong W, Zhang JH. Melatonin decreases mortality following severe subarachnoid hemorrhage. Journal of Pineal Research. 2008;44:197-204.
  225. Zausinger S, Westermaier T, Plesnila N, Steiger HJ, Schmid-Elsaesser R. Neuroprotection in Transient Focal Cerebral Ischemia by Combination Drug Therapy and Mild Hypothermia: Comparison With Customary Therapeutic Regimen. Stroke. 2003;34:1526-1532.
Copyright © 2014 Journal of Pharmacopuncture. All rights reserved.