The Acid Sphingomyelinase/ Ceramide System as Target for Ischemic Stroke Therapies

 

Ayan Mohamud Yusuf    Nina Hagemann    Dirk M. Hermann

 

Department of Neurology, University Hospital Essen, Essen, Germany

 

 

 

 

Key Words

Stroke • Acid sphingomyelinase • Ceramide

 

Abstract

In this review, we summarize implications of the acid sphingomyelinase/ ceramide system in ischemic stroke. Acid sphingomyelinase catalyzes the formation of the bioactive sphingolipid ceramide which coalesces into membrane platforms and has a pivotal role in inflammation, cell signaling and death. Cerebral ischemia increases acid sphingomyelinase activity and elevates brain ceramide levels, which has been associated with the exacerbation of ischemic injury and deterioration of stroke outcome. In view of the fact that lowering acid sphingomyelinase activity and ceramide level was shown to protect against ischemic injury and ameliorate neurological deficits, the acid sphingomyelinase/ ceramide system might represent a promising target for stroke therapies.

 

 

Stroke

 

Background

Ischemic stroke, defined as an acute neurological deficit caused by the thromboembolic occlusion of a brain-supplying artery, is a leading cause of death and disability among adults worldwide [1]. The available treatment strategies for acute ischemic stroke (AIS) aim for recanalization of the occluded vessel by means of systemic thrombolysis and/ or mechanical thrombectomy. Recombinant tissue-plasminogen activator (rtPA) is a thrombolytic which is delivered intravenously (i.v) and the only pharmacological intervention approved by the US Food and Drug Administration (FDA) in 1996 after rtPA treatment was shown to reduce disability and mortality when delivered within up to 4.5 hours following stroke [2, 3]. Because of this short time window, only a small percentage of stroke patients (up to 30 percent, depending on health care system) benefits from intravenous thrombolysis. Recanalization can also be achieved by endovascular mechanical thrombectomy with a stent retriever which is indicated in case of proximal large vessel occlusions [4]. The first device used for this procedure received FDA approval in 2004 [5]. Thrombectomy is mostly performed in addition to thrombolysis [6]. According to American Heart Association/American Stroke Association guidelines from 2018 this procedure is recommended within 6 hours after onset of stroke symptoms [7]. Both intravenous thrombolysis and mechanical thrombectomy may induce side effects, such as brain hemorrhages [8, 9]. There is a clear need for treatments that allow promoting stroke recovery when the time window for acute treatments has exceeded.

 

Pathophysiology of ischemic stroke

The occlusion of a brain-supplying artery results in focal cerebral ischemia. Focal cerebral ischemia is characterized by a central core region with strongly compromised cerebral blood flow (CBF) that is surrounded by the so called penumbra, an area that is still viable and has a CBF that is below functional thresholds [10].

Major pathological events that occur during cerebral ischemia are massive excitatory neurotransmitter release (specifically of glutamate) associated with peri-infarct depolarizations occurring within minutes, inflammatory responses peaking after 24 hours to a few days and delayed neuronal injury that is most prominent in the first days but progresses over weeks resulting in secondary neurodegeneration and brain atrophy [11].

These pathophysiological events are directly initiated by the cerebral hypoperfusion. The impaired delivery of glucose and oxygen to the brain leads to mitochondrial dysfunction and a reduced synthesis of adenosine triphosphate (ATP) [12]. The depletion of ATP causes an impaired function of Na+/K+-ATPases. Consequently, neurons and glia cells depolarize and excitatory amino acids such as glutamate are released which accumulate in the synaptic space since uptake is also compromised, leading to an overactivation of N-methyl-D-aspartate (NMDA) receptors and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Further, as Na+ and Cl- enter the cell, water follows passively which leads to cell swelling. The overactivation of glutamate receptors induces a cellular calcium influx that triggers the activation of calcium-dependent proteases, endonucleases and lipases [13]. The calcium-mediated activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase catalyzes superoxide production which mediates cell death [14]. The release of damage-associated molecular pattern (DAMP) molecules from dying neurons activate resident microglia that produce inflammatory cytokines such as the tumor necrosis factor α (TNF-α), interleukin 1 (IL-1) and interleukin 6 (IL-6) [15-20]. The expression of adhesion molecules such as the intercellular adhesion molecule 1 (ICAM-1) on cerebral endothelial cells is increased allowing the infiltration of inflammatory cells such as neutrophils that further exacerbate brain damage [21, 22]. Further, the matrix metalloproteinase 9 (MMP9) disrupts the integrity of the blood-brain-barrier (BBB) by degradation of the basal lamina and tight junctions, consequently leading to edema formation and possibly hemorrhagic transformation [23-27].

 

Failed strategies for ischemic stroke treatment

Despite extensive research and promising data from pre-clinical stroke model, a plethora of supposedly neuroprotective compounds targeting specific elements of the ischemic cascade have failed in clinical trials. For reasons of space, we only touch a few examples here. Several trials assessed the potential benefit of calcium antagonists after preclinical data indicated that blocking of calcium channels reduces infarct size and brain edema [28, 29]. The Very Early Nimodipine Use in Stroke (VENUS) trial has approached this with nimodipine, a calcium channel blocker that is used to prevent the occurrence of subarachnoid hemorrhage (SAH) related vasospasms. The VENUS trial enrolled 454 patients, which were treated within 6 hours after stroke onset. No differences were observed between the treated group and the placebo group at trial termination [30]. Likewise, the Flunarizine in Stroke Treatment (FIST) trial analyzed the effect of the calcium channel blocker flunarizine, which is clinically used for treating migraine. 331 patients were enrolled and treatment started within 24 hours after stroke onset. Flunarizine was not superior to placebo in this study [31]. Other studies similarly evaluated nimodipine or flunarizine, all with lack of clinical benefits [32].

NMDA receptors antagonists have been designed because of the high glutamate concentrations released in ischemic tissue that exacerbate brain injury [33-35]. The efficacy of the NMDA antagonist aptiganel hydrochloride was investigated in a trial with 628 patients who were treated within 6 hours after stroke. The treatment was not efficacious and due to cognitive side effects raised serious safety concerns [36]. The Glycine Antagonist in Neuroprotection (GAIN) trial examined the efficacy of another NMDA antagonist called gavestinel and enrolled 1367 patients that were treated within 6 hours after symptom onset. Also, gavestinel failed to alleviate functional outcome [37], as did studies with other NMDA receptor antagonists [32].

Since inflammation is a crucial element in stroke pathology, efforts have been made to attenuate the inflammatory response after stroke. Based on observations that mice deficient for ICAM-1 were protected from ischemia-reperfusion injury [38], a monoclonal murine ICAM-1 antibody called enlimomab was tested in 625 patients who received the antibody or placebo within 6 hours after stroke onset. Enlimomab was not effective, but was associated with an elevated mortality rate [39]. Another attempt to dampen the inflammatory response after stroke was the delivery of a humanized antibody called rovelizumab which targets the β2-subunit of the lymphocyte function–associated antigen-1 (LFA-1) and macrophage-1 antigen (Mac-1) which bind to ICAM-1. The antibody was well tolerated but the trial was halted at an interim analysis because a beneficial effect could not be detected [40]. In the meantime, concerns have been raised about the mouse mutants used for examining effects of ICAM-1 knockout. While membrane bound ICAM-1 is not detectable, soluble forms of ICAM-1 are still present in the serum of these mice [41]. Based on these insights, ICAM-1 null mice with a deletion of the entire coding region of ICAM-1 have been developed, which did not reveal any beneficial effects of ICAM-1 deficiency in ischemic stroke models. Specifically, there was no difference in infarct size compared to wildtype mice and the disruption of the blood-brain-barrier was even more pronounced in ICAM-1 null than wildtype mice [42].

Additionally, a large variety of neuroprotective agents acting including free radical scavengers, nitric oxide inhibitors, AMPA antagonists, GABA agonists, sodium channel blockers or gangliosides have been studied in clinical stroke trials, all without benefits [43].

 

 

Acid sphingomyelinase/ ceramide system

 

Synthesis and structure of the acid sphingomyelinase

Acid sphingomyelinase (ASM) is a lysosomal phosphodiesterase which is encoded by the sphingomyelin phosphodiesterase gene (SMPD1) that is expressed on chromosome 11p15.1-p15.4 [44].

ASM is synthesized as an N-glycosylated 75 kDa pre-pro-enzyme, which is targeted to the endoplasmic reticulum (ER) where the N-terminal signal peptide is cleaved which leads to the formation of the 72 kDa pro-enzyme [45, 46]. This precursor is post-translationally subjected to glycosylation inside the Golgi complex [47]. Lysosomal ASM (L-ASM) acquires high mannose oligosaccharides, whereas secretory ASM (S-ASM) has a more complex glycosylation composition [45, 48]. L-ASM is targeted to the lysosome via mannose 6-phosphate receptor or sortilin-mediated pathways and binds Zn2+ ions during this trafficking process [45, 48, 49]. It is further proteolytically processed at the C-terminus which is necessary for the enzyme to obtain its catalytic activity [50]. S-ASM is released from the cell via the Golgi secretory pathway and additionally requires extracellular Zn2+ for its activation [48]. The crystal structure of mature ASM revealed that the enzyme is composed of an N-terminal saposin domain, a C-terminal metallophosphoesterase catalytic domain with two Zn2+ ions and a prolin-rich connector domain [51, 52]. Sphingomyelin binds to ASM by positioning its ceramide-phosphate group at the Zn2+ center [51]. The catalytic activity of ASM is dependent on an acidic pH and its attachment to membrane surfaces that is mediated by electrostatic forces [51].

 

Functional inhibitors of ASM (FIASMAs)

Several antidepressants such as amitriptyline, fluoxetine and nortriptyline are functional inhibitors of ASM (FIASMAs) [53]. Antidepressants were long believed to act via inhibition of monoamine reuptake. While fluoxetine is a selective serotonin reuptake inhibitor, amitriptyline preferentially inhibits serotonin and noradrenalin reuptake and nortriptyline preferentially inhibits noradrenalin reuptake [54]. However, tianeptine is a serotonin reuptake enhancer [55]. The monoamine mechanism for the induction of antidepressant effects was challenged by observations that amitriptyline and fluoxetine reduced ASM activity and ceramide concentration, inducing neurogenesis and behavioral recovery in a mouse model of depression in wildtype mice but not ASM deficient mice [56]. These experiments clearly identified a critical role of the ASM/ ceramide system as target for antidepressants.

Despite their structural heterogeneity, FIASMAs have lipophilic and weakly basic properties in common [53]. As lysosomotropic compounds, FIASMAs passively enter the lysosome through the lysosomal membrane, become protonated in this acidic compartment, accumulate and interfere with the attachment of ASM to the lysosomal membrane, consequently leading to a proteolytic degradation of ASM by lysosomal proteases [53, 57-59]. Treatment with FIASMAs does not induce complete ASM degradation [56, 60]. It is not clear yet if the remaining ASM activity originates from lysosomes but a basal ASM activity might be necessary to prevent pathologies resembling Niemann-Pick disease that is caused by genetic ASM deficiency [53, 61]. The simultaneous treatment with multiple FIASMAs results in an amplified inhibition of ASM [62].

FIASMAs do not generally abrogate the activity of all lysosomal hydrolases. Yet, desipramine, chloroquine and chlorpromazine were also shown to inhibit the lysosomal enzymes acid ceramidase (AC), acid lipase and phospholipases A and C [53]. Since FIASMAs are capable of passively diffusing through the blood-brain barrier (BBB) [62], they are attractive tools not only in the treatment of depression, but potentially also of ischemic and degenerative brain disease.

 

 

ASM and ceramide signaling in experimental ischemic stroke

 

The sphingomyelinase pathway is activated by stress stimuli for instance in response to irradiation, acute systemic inflammation induced by lipopolysaccharide (LPS) or upon release of reactive oxygen species (ROS) or pro-inflammatory cytokines such as the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) [63-67]. Upon activation, ASM is translocated from the lysosomes to the extracellular leaflet of the cell membrane, bringing it in close proximity to its substrate sphingomyelin [68, 69]. ASM then hydrolyzes sphingomyelin producing ceramide and phosphorylcholine. Due to its biophysical properties ceramide coalesces into microdomains that fuse and form large ceramide-enriched membrane platforms with a diameter of 200 nanometer up to several micrometer [70]. Ceramide-enriched membrane platforms induce the spatial reorganisation and clustering of membrane proteins and receptors, leading to the amplification of their elicited cell signals [71]. Ceramide-rich membrane platforms are critically involved in induction of apoptosis and growth inhibition, besides others [70].

Experimental studies provided evidence for the significance of the ASM/ ceramide system in ischemic stroke (Table 1). In mice subjected to 60 minutes transient proximal middle cerebral artery occlusion (MCAO), ASM activity and ceramide level were significantly increased in the ischemic cerebral cortex after 6 hours reperfusion [72]. Ceramide was not increased upon MCAO in ASM knockout mice [72]. ASM deficiency reduced infarct size, improved neurological deficits and inhibited the production of inflammatory cytokines [72]. Primary cortical neuronal cultures from ASM knockout mice exhibited less vulnerability to glutamate-induced toxicity [72]. Intracerebroventricular administration of xanthogenate tricyclodecan-9-yl (D609), an inhibitor of the phosphatidylcholine (PC)-specific phospholipase C that catalyzes the formation of ASM-activating diacylglycerol (DAG) [73, 74], 30 minutes before MCAO and 2 hours after reperfusion prevented the increase of ASM activity and ceramide level, dampened pro-inflammatory cytokine production, attenuated neuronal damage and improved post-stroke behavior [72]. Interestingly, the ceramide increase in ischemic stroke is strictly linked to reperfusion. Thus, ischemic mice revealed this increased ceramide level in ischemic tissue only when transient (30 minutes) proximal but not permanent proximal MCAO was induced [75]. Yu et al. did not observe any alterations in sphingomyelin content, thus excluding sphingomyelin hydrolysis as the ceramide source in this case of mild focal cerebral ischemia and postulating that ceramide was rather generated by de novo biosynthesis because intermediate products of this alternative pathway for ceramide generation such as dihydrosphingosine and dihydroceramide were increased [75]. In rats exposed to 90 minutes transient proximal MCAO, ceramide levels increased in the thalamus by 190% and in the entorhinal cortex by 175% after 6 hours reperfusion, which was prevented when FK506 was administered 5 minutes after MCAO induction [76]. Complementary findings were made in an in vitro model of ischemia. Human neuroblastoma cells exposed to low oxygen concentrations and serum starvation revealed increased ASM activity and ceramide level after 30 minutes of reoxygenation under conditions resulting in apoptotic cell injury [76]. Preincubation with FK506 counteracted the ceramide increase and apoptosis induction [76]. FK506 is neuroprotective in several experimental stroke models [77-79]. FK506 has prominent immunomodulatory effects [80]. In the ischemic brain, FK506 was shown to downregulate pro-inflammatory cytokines and dampen the activation of astrocytes [81, 82]. In retinal ischemia, FK506 reduced leukocyte accumulation [83]. In a model of chronic bilateral cerebral ischemia induced by bilateral carotid artery occlusion (BCAO) in rats, ASM activity and ceramide level were unchanged at day 1 after BCAO and significantly increased at day 3, 7 and 14 in the frontal cortex, corpus callosum, internal capsule and globus pallidus [84]. Ceramide was found in glial fibrillary acidic protein (GFAP)-positive astroglia [84]. An increase of ceramide and a decrease of sphingomyelin was noted in the hippocampus 30 minutes and 24 hours after 5 minutes of BCAO in gerbils [85], indicating that ceramide was produced via the sphingomyelinase pathway. Ceramide accumulation depends on the severity of the ischemic episode. No ceramide changes were observed after 2 minutes of BCAO [85].

 

Table 1. ASM and ceramide signaling in experimental ischemic stroke

 

 

Effects of ASM inhibitors in experimental ischemic stroke

 

Experimental studies have also investigated the utility of FIASMAs in stroke treatment (Table 2). In rats exposed to 60 minutes transient proximal MCAO, fluoxetine reduced infarct volume and ameliorated motor deficits [86]. A low dose of fluoxetine (1 mg/kg) was neuroprotective when administered (i.v.) at 30 minutes post-stroke, but at 9 hours post-stroke a high dose (10 mg/kg) was necessary for the induction of tissue survival and motor recovery [86]. Even more delayed treatment after 12 did not confer neuroprotection or enhance motor deficits [86]. In rats treated with fluoxetine at 5 mg/kg or 10 mg/kg (i.v.) 6 hours post-stroke, inflammatory responses were reduced, demonstrated by a decrease in the activation of the nuclear factor kappa light-chain enhancer of activated B-cells (NF-κB), a reduction in microglia activation and a reduction in brain neutrophil infiltration [86]. The authors confirmed the anti-inflammatory effects of fluoxetine in primary microglial cells or primary neutrophils treated with lipopolysaccharide (LPS) and reported a fluoxetine-mediated reduction in NF-κB activation and pro-inflammatory cytokine gene expression levels [86]. Post-ischemic hyperactivity in gerbils was reduced by pretreatment with fluoxetine intraperitoneally (i.p.) administered at doses of 10 mg/kg, 20 mg/kg or 40 mg/kg 30 minutes before 5 minutes of BCAO [87]. High dose fluoxetine pretreatment at 40 mg/kg (i.p.) for 3 consecutive days before BCAO also increased neuronal survival in the CA1 hippocampal area [87]. Mice receiving fluoxetine in the drinking water (120 mg/l) from day 3 to day 28 after photothrombotic stroke revealed a reduction in final stroke size [88]. In the peri-infarct cortex, the ceramide level was increased at day 3 and 7 post-stroke due to de novo synthesis and not ASM activation [88]. In this study, fluoxetine did not improve neurological recovery [88]. After focal cerebral ischemia in rats induced by endothelin-1 injection fluoxetine delivery via miniosmotic pumps (10 mg/kg) for 4 weeks starting on day 7 did not enhance motor function [89]. Neurological outcome and infarct volume were not altered in rats exposed to 120 minutes transient proximal MCAO that were treated with fluoxetine at 5 mg/kg (i.p.) for 10 days from day 2 [90].

 

Table 2. Effects of ASM inhibitors in experimental ischemic stroke

 

 

Effects of ASM inhibitors in clinical stroke trials

 

Due to the lack of safety concerns, ASM inhibitors have been studied in clinical trials. In a placebo-controlled study in ischemic stroke patients exhibiting hemiparesis or hemiplegia, fluoxetine (20 mg/day) initiated between day 5 and day 10 post-stroke and continued for 90 days in addition to physiotherapy enhanced motor recovery [91], suggesting that fluoxetine (20 mg/day) modulates brain plasticity. Indeed, another study in patients with hemiparesis showed that even a single dose of fluoxetine (20 mg) 14 days after stroke modulated cerebral motor activation and enhanced hand motor function [92]. The effect on motor function after a single bolus injection excludes that motor effects are attributed to the enhancement of the patients’ mood that normally requires a longer treatment time [91]. Further, treatment with fluoxetine (10 mg/day gradually increased to 40 mg/day) or nortriptyline (25 mg/day gradually increased to 100 mg/day) for 12 weeks within the first 6 months after stroke increased the probability of survival irrespective of whether the patient suffered from depression at enrollment or not [93]. In patients with stroke-associated neuropathic pain without depressive symptoms, 4 weeks of amitriptyline treatment (25 mg/day gradually increased to 75 mg/day) induced pain relief [94].

There have also been studies lacking recovery promoting effects. In stroke patients with persistent neurological deficits treated with placebo or fluoxetine (20 mg/day) starting 2 days to 15 days post-stroke for 6 months, fluoxetine reduced the incidence of depression but increased the incidence of bone fractures [95]. Importantly, fluoxetine did not influence neurological outome or stroke survival [95]. A study comparing the efficacy of fluoxetine (10 mg/day gradually increased to 40 mg/day) or nortriptyline (25 mg/day gradually increased to 100 mg/day) for 12 weeks in depressed and non-depressed patients that had a stroke within 6 months before, found that nortriptyline ameliorated post-stroke depression, whereas both antidepressants did not enhance stroke-related physical and cognitive impairments [96].

 

 

Conclusion

 

There is compelling evidence that the ASM/ ceramide system is critically involved in ischemic stroke pathogenesis. A variety of experimental studies demonstrated beneficial effects of ASM inhibition and ceramide lowering for ischemic brain injury. Solid evidence was obtained that FIASMAs reduce ischemic injury in the acute stroke phase and in addition experimental studies suggested that ASM inhibition may also induce neurological recovery in the post-acute stroke phase. Unfortunately, these studies so far did not elucidate whether these restorative actions were specifically attributed to the modulation of the ASM/ ceramide system. Further studies are needed to address this question.

FIASMAs have been used in clinics for decades which provides the advantage that possible side effects, toxicity and contraindications are well known. Due to their broad mechanisms of action, it at this stage remains speculative to assign recovery promoting actions in stroke patients to their inhibitory effect on ASM. Specific ASM inhibitors with negligible off-target effects are not available for use in humans so there is currently no alternative to achieve ASM inhibition in patients other than by FIASMAs [53]. Since clinical trials using FIASMAs were inconsistent, a generalized statement on the efficacy of FIASMAs in stroke patients cannot be made.

 

 

Disclosure Statement

 

The authors declare that they have no conflicts of interest.

 

 

References

 

1 Katan M, Luft A: Global Burden of Stroke. Semin Neurol 2018;38:208-211.
https://doi.org/10.1055/s-0038-1649503

 

2 Hacke W, Kaste M, Bluhmki E, Brozman M, Davalos A, Guidetti D, Larrue V, Lees KR, Medeghri Z, Machnig T, Schneider D, von Kummer R, Wahlgren N, Toni D, Investigators E: Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J Med 2008;359:1317-1329.
https://doi.org/10.1056/NEJMoa0804656

 

3 National Institute of Neurological Disorders: Tissue plasminogen activator for acute ischemic stroke. N Engl J Med 1995;333:1581-1587.
https://doi.org/10.1056/NEJM199512143332401

 

4 Vidale S, Longoni M, Valvassori L, Agostoni E: Mechanical Thrombectomy in Strokes with Large-Vessel Occlusion Beyond 6 Hours: A Pooled Analysis of Randomized Trials. J Clin Neurol 2018;14:407-412.
https://doi.org/10.3988/jcn.2018.14.3.407

 

5 Katz JM, Gobin YP: Merci Retriever in acute stroke treatment. Expert Rev Med Devices 2006;3:273-280.
https://doi.org/10.1586/17434440.3.3.273

 

6 Campbell BCV, Donnan GA, Lees KR, Hacke W, Khatri P, Hill MD, Goyal M, Mitchell PJ, Saver JL, Diener HC, Davis SM: Endovascular stent thrombectomy: the new standard of care for large vessel ischaemic stroke. Lancet Neurol 2015;14:846-854.
https://doi.org/10.1016/S1474-4422(15)00140-4

 

7 Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B, Jauch EC, Kidwell CS, Leslie-Mazwi TM, Ovbiagele B, Scott PA, Sheth KN, Southerland AM, Summers DV, Tirschwell DL, American Heart Association Stroke C: 2018 Guidelines for the Early Management of Patients With Acute Ischemic Stroke: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke 2018;49:e46-e110.
https://doi.org/10.1161/STR.0000000000000211

 

8 Balami JS, White PM, McMeekin PJ, Ford GA, Buchan AM: Complications of endovascular treatment for acute ischemic stroke: Prevention and management. Int J Stroke 2018;13:348-361.
https://doi.org/10.1177/1747493017743051

 

9 Clark WM, Albers GW, Madden KP, Hamilton S: The rtPA (alteplase) 0- to 6-hour acute stroke trial, part A (A0276g) : results of a double-blind, placebo-controlled, multicenter study. Thromblytic therapy in acute ischemic stroke study investigators. Stroke 2000;31:811-816.
https://doi.org/10.1161/01.STR.31.4.811

 

10 Hata R, Maeda K, Hermann D, Mies G, Hossmann KA: Dynamics of regional brain metabolism and gene expression after middle cerebral artery occlusion in mice. J Cereb Blood Flow Metab 2000;20:306-315.
https://doi.org/10.1097/00004647-200002000-00012

 

11 Hermann DM, Chopp M: Promoting brain remodelling and plasticity for stroke recovery: therapeutic promise and potential pitfalls of clinical translation. Lancet Neurol 2012;11:369-380.
https://doi.org/10.1016/S1474-4422(12)70039-X

 

12 Song M, Yu SP: Ionic regulation of cell volume changes and cell death after ischemic stroke. Transl Stroke Res 2014;5:17-27.
https://doi.org/10.1007/s12975-013-0314-x

 

13 Lipton P: Ischemic cell death in brain neurons. Physiol Rev 1999;79:1431-1568.
https://doi.org/10.1152/physrev.1999.79.4.1431

 

14 Brennan AM, Suh SW, Won SJ, Narasimhan P, Kauppinen TM, Lee H, Edling Y, Chan PH, Swanson RA: NADPH oxidase is the primary source of superoxide induced by NMDA receptor activation. Nat Neurosci 2009;12:857-863.
https://doi.org/10.1038/nn.2334

 

15 Zaremba J, Skrobanski P, Losy J: Tumour necrosis factor-alpha is increased in the cerebrospinal fluid and serum of ischaemic stroke patients and correlates with the volume of evolving brain infarct. Biomed Pharmacother 2001;55:258-263.
https://doi.org/10.1016/S0753-3322(01)00058-0

 

16 Perini F, Morra M, Alecci M, Galloni E, Marchi M, Toso V: Temporal profile of serum anti-inflammatory and pro-inflammatory interleukins in acute ischemic stroke patients. Neurol Sci 2001;22:289-296.
https://doi.org/10.1007/s10072-001-8170-y

 

17 Luheshi NM, Kovacs KJ, Lopez-Castejon G, Brough D, Denes A: Interleukin-1alpha expression precedes IL-1beta after ischemic brain injury and is localised to areas of focal neuronal loss and penumbral tissues. J Neuroinflammation 2011;8:186.
https://doi.org/10.1186/1742-2094-8-186

 

18 Clausen BH, Lambertsen KL, Babcock AA, Holm TH, Dagnaes-Hansen F, Finsen B: Interleukin-1beta and tumor necrosis factor-alpha are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J Neuroinflammation 2008;5:46.
https://doi.org/10.1186/1742-2094-5-46

 

19 Suzuki S, Tanaka K, Nogawa S, Nagata E, Ito D, Dembo T, Fukuuchi Y: Temporal profile and cellular localization of interleukin-6 protein after focal cerebral ischemia in rats. J Cereb Blood Flow Metab 1999;19:1256-1262.
https://doi.org/10.1097/00004647-199911000-00010

 

20 Kanazawa M, Ninomiya I, Hatakeyama M, Takahashi T, Shimohata T: Microglia and Monocytes/Macrophages Polarization Reveal Novel Therapeutic Mechanism against Stroke. Int J Mol Sci 2017;18:pii:E2135.
https://doi.org/10.3390/ijms18102135

 

21 Stanimirovic D, Shapiro A, Wong J, Hutchison J, Durkin J: The induction of ICAM-1 in human cerebromicrovascular endothelial cells (HCEC) by ischemia-like conditions promotes enhanced neutrophil/HCEC adhesion. J Neuroimmunol 1997;76:193-205.
https://doi.org/10.1016/S0165-5728(97)00057-X

 

22 Neumann J, Riek-Burchardt M, Herz J, Doeppner TR, Konig R, Hutten H, Etemire E, Mann L, Klingberg A, Fischer T, Gortler MW, Heinze HJ, Reichardt P, Schraven B, Hermann DM, Reymann KG, Gunzer M: Very-late-antigen-4 (VLA-4)-mediated brain invasion by neutrophils leads to interactions with microglia, increased ischemic injury and impaired behavior in experimental stroke. Acta Neuropathol 2015;129:259-277.
https://doi.org/10.1007/s00401-014-1355-2

 

23 Clark AW, Krekoski CA, Bou SS, Chapman KR, Edwards DR: Increased gelatinase A (MMP-2) and gelatinase B (MMP-9) activities in human brain after focal ischemia. Neurosci Lett 1997;238:53-56.
https://doi.org/10.1016/S0304-3940(97)00859-8

 

24 Ramos-Fernandez M, Bellolio MF, Stead LG: Matrix metalloproteinase-9 as a marker for acute ischemic stroke: a systematic review. J Stroke Cerebrovasc Dis 2011;20:47-54.
https://doi.org/10.1016/j.jstrokecerebrovasdis.2009.10.008

 

25 Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, Davalos A: Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke. Stroke 2003;34:40-46.
https://doi.org/10.1161/01.STR.0000046764.57344.31

 

26 Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA: Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab 2007;27:697-709.
https://doi.org/10.1038/sj.jcbfm.9600375

 

27 Rosenberg GA, Yang Y: Vasogenic edema due to tight junction disruption by matrix metalloproteinases in cerebral ischemia. Neurosurg Focus 2007;22:E4.
https://doi.org/10.3171/foc.2007.22.5.5

 

28 Korenkov AI, Pahnke J, Frei K, Warzok R, Schroeder HW, Frick R, Muljana L, Piek J, Yonekawa Y, Gaab MR: Treatment with nimodipine or mannitol reduces programmed cell death and infarct size following focal cerebral ischemia. Neurosurg Rev 2000;23:145-150.
https://doi.org/10.1007/PL00011946

 

29 Deng W, Feng Y: Effect of dl-3-n-butylphthalide on brain edema in rats subjected to focal cerebral ischemia. Chin Med Sci J 1997;12:102-106.

 

30 Horn J, de Haan RJ, Vermeulen M, Limburg M: Very Early Nimodipine Use in Stroke (VENUS): a randomized, double-blind, placebo-controlled trial. Stroke 2001;32:461-465.
https://doi.org/10.1161/01.STR.32.2.461

 

31 Franke CL, Palm R, Dalby M, Schoonderwaldt HC, Hantson L, Eriksson B, Lang-Jenssen L, Smakman J: Flunarizine in stroke treatment (FIST): a double-blind, placebo-controlled trial in Scandinavia and the Netherlands. Acta Neurol Scand 1996;93:56-60.
https://doi.org/10.1111/j.1600-0404.1996.tb00171.x

 

32 Xu SY, Pan SY: The failure of animal models of neuroprotection in acute ischemic stroke to translate to clinical efficacy. Med Sci Monit Basic Res 2013;19:37-45.
https://doi.org/10.12659/MSMBR.883750

 

33 Choi DW: Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett 1985;58:293-297.
https://doi.org/10.1016/0304-3940(85)90069-2

 

34 Simon RP, Swan JH, Griffiths T, Meldrum BS: Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain. Science 1984;226:850-852.
https://doi.org/10.1126/science.6093256

 

35 Takano K, Tatlisumak T, Formato JE, Carano RA, Bergmann AG, Pullan LM, Bare TM, Sotak CH, Fisher M: Glycine site antagonist attenuates infarct size in experimental focal ischemia. Postmortem and diffusion mapping studies. Stroke 1997;28:1255-1263.
https://doi.org/10.1161/01.STR.28.6.1255

 

36 Albers GW, Goldstein LB, Hall D, Lesko LM, Aptiganel Acute Stroke I: Aptiganel hydrochloride in acute ischemic stroke: a randomized controlled trial. JAMA 2001;286:2673-2682.
https://doi.org/10.1001/jama.286.21.2673

 

37 Sacco RL, DeRosa JT, Haley EC, Jr., Levin B, Ordronneau P, Phillips SJ, Rundek T, Snipes RG, Thompson JL, Glycine Antagonist in Neuroprotection Americas I: Glycine antagonist in neuroprotection for patients with acute stroke: GAIN Americas: a randomized controlled trial. JAMA 2001;285:1719-1728.
https://doi.org/10.1001/jama.285.13.1719

 

38 Connolly ES, Jr., Winfree CJ, Springer TA, Naka Y, Liao H, Yan SD, Stern DM, Solomon RA, Gutierrez-Ramos JC, Pinsky DJ: Cerebral protection in homozygous null ICAM-1 mice after middle cerebral artery occlusion. Role of neutrophil adhesion in the pathogenesis of stroke. J Clin Invest 1996;97:209-216.
https://doi.org/10.1172/JCI118392

 

39 Enlimomab Acute Stroke Trial I: Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology 2001;57:1428-1434.
https://doi.org/10.1212/WNL.57.8.1428

 

40 Becker KJ: Anti-leukocyte antibodies: LeukArrest (Hu23F2G) and Enlimomab (R6.5) in acute stroke. Curr Med Res Opin 2002;18:s18-22.
https://doi.org/10.1185/030079902125000688

 

41 van Den Engel NK, Heidenthal E, Vinke A, Kolb H, Martin S: Circulating forms of intercellular adhesion molecule (ICAM)-1 in mice lacking membranous ICAM-1. Blood 2000;95:1350-1355.
https://doi.org/10.1182/blood.V95.4.1350.004k07_1350_1355

 

42 Enzmann GU, Pavlidou S, Vaas M, Klohs J, Engelhardt B: ICAM-1(null) C57BL/6 Mice Are Not Protected from Experimental Ischemic Stroke. Transl Stroke Res 2018;9:608-621.
https://doi.org/10.1007/s12975-018-0612-4

 

43 O'Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW: 1, 026 experimental treatments in acute stroke. Ann Neurol 2006;59:467-477.
https://doi.org/10.1002/ana.20741

 

44 Schuchman EH, Levran O, Pereira LV, Desnick RJ: Structural organization and complete nucleotide sequence of the gene encoding human acid sphingomyelinase (SMPD1). Genomics 1992;12:197-205.
https://doi.org/10.1016/0888-7543(92)90366-Z

 

45 Hurwitz R, Ferlinz K, Vielhaber G, Moczall H, Sandhoff K: Processing of human acid sphingomyelinase in normal and I-cell fibroblasts. J Biol Chem 1994;269:5440-5445.

 

46 Wan Q, Schuchman EH: A novel polymorphism in the human acid sphingomyelinase gene due to size variation of the signal peptide region. Biochim Biophys Acta 1995;1270:207-210.
https://doi.org/10.1016/0925-4439(95)00050-E

 

47 Newrzella D, Stoffel W: Functional analysis of the glycosylation of murine acid sphingomyelinase. J Biol Chem 1996;271:32089-32095.
https://doi.org/10.1074/jbc.271.50.32089

 

48 Schissel SL, Keesler GA, Schuchman EH, Williams KJ, Tabas I: The cellular trafficking and zinc dependence of secretory and lysosomal sphingomyelinase, two products of the acid sphingomyelinase gene. J Biol Chem 1998;273:18250-18259.
https://doi.org/10.1074/jbc.273.29.18250

 

49 Ni X, Morales CR: The lysosomal trafficking of acid sphingomyelinase is mediated by sortilin and mannose 6-phosphate receptor. Traffic 2006;7:889-902.
https://doi.org/10.1111/j.1600-0854.2006.00429.x

 

50 Jenkins RW, Idkowiak-Baldys J, Simbari F, Canals D, Roddy P, Riner CD, Clarke CJ, Hannun YA: A novel mechanism of lysosomal acid sphingomyelinase maturation: requirement for carboxyl-terminal proteolytic processing. J Biol Chem 2011;286:3777-3788.
https://doi.org/10.1074/jbc.M110.155234

 

51 Xiong ZJ, Huang J, Poda G, Pomes R, Prive GG: Structure of Human Acid Sphingomyelinase Reveals the Role of the Saposin Domain in Activating Substrate Hydrolysis. J Mol Biol 2016;428:3026-3042.
https://doi.org/10.1016/j.jmb.2016.06.012

 

52 Gorelik A, Illes K, Heinz LX, Superti-Furga G, Nagar B: Crystal structure of mammalian acid sphingomyelinase. Nat Commun 2016;7:12196.
https://doi.org/10.1038/ncomms12196

 

53 Kornhuber J, Tripal P, Reichel M, Muhle C, Rhein C, Muehlbacher M, Groemer TW, Gulbins E: Functional Inhibitors of Acid Sphingomyelinase (FIASMAs): a novel pharmacological group of drugs with broad clinical applications. Cell Physiol Biochem 2010;26:9-20.
https://doi.org/10.1159/000315101

 

54 Sanchez C, Hyttel J: Comparison of the effects of antidepressants and their metabolites on reuptake of biogenic amines and on receptor binding. Cell Mol Neurobiol 1999;19:467-489.
https://doi.org/10.1023/A:1006986824213

 

55 Brink CB, Harvey BH, Brand L: Tianeptine: a novel atypical antidepressant that may provide new insights into the biomolecular basis of depression. Recent Pat CNS Drug Discov 2006;1:29-41.
https://doi.org/10.2174/157488906775245327

 

56 Gulbins E, Palmada M, Reichel M, Luth A, Bohmer C, Amato D, Muller CP, Tischbirek CH, Groemer TW, Tabatabai G, Becker KA, Tripal P, Staedtler S, Ackermann TF, van Brederode J, Alzheimer C, Weller M, Lang UE, Kleuser B, Grassme H, Kornhuber J: Acid sphingomyelinase-ceramide system mediates effects of antidepressant drugs. Nat Med 2013;19:934-938.
https://doi.org/10.1038/nm.3214

 

57 Kolzer M, Werth N, Sandhoff K: Interactions of acid sphingomyelinase and lipid bilayers in the presence of the tricyclic antidepressant desipramine. FEBS Lett 2004;559:96-98.
https://doi.org/10.1016/S0014-5793(04)00033-X

 

58 Daniel WA, Wojcikowski J: Contribution of lysosomal trapping to the total tissue uptake of psychotropic drugs. Pharmacol Toxicol 1997;80:62-68.
https://doi.org/10.1111/j.1600-0773.1997.tb00285.x

 

59 Hurwitz R, Ferlinz K, Sandhoff K: The tricyclic antidepressant desipramine causes proteolytic degradation of lysosomal sphingomyelinase in human fibroblasts. Biol Chem Hoppe Seyler 1994;375:447-450.
https://doi.org/10.1515/bchm3.1994.375.7.447

 

60 Kornhuber J, Medlin A, Bleich S, Jendrossek V, Henkel AW, Wiltfang J, Gulbins E: High activity of acid sphingomyelinase in major depression. J Neural Transm (Vienna) 2005;112:1583-1590.
https://doi.org/10.1007/s00702-005-0374-5

 

61 Brady RO, Kanfer JN, Mock MB, Fredrickson DS: The metabolism of sphingomyelin. II. Evidence of an enzymatic deficiency in Niemann-Pick diseae. Proc Natl Acad Sci U S A 1966;55:366-369.
https://doi.org/10.1073/pnas.55.2.366

 

62 Kornhuber J, Muehlbacher M, Trapp S, Pechmann S, Friedl A, Reichel M, Muhle C, Terfloth L, Groemer TW, Spitzer GM, Liedl KR, Gulbins E, Tripal P: Identification of novel functional inhibitors of acid sphingomyelinase. PLoS One 2011;6:e23852.
https://doi.org/10.1371/journal.pone.0023852

 

63 Wong ML, Xie B, Beatini N, Phu P, Marathe S, Johns A, Gold PW, Hirsch E, Williams KJ, Licinio J, Tabas I: Acute systemic inflammation up-regulates secretory sphingomyelinase in vivo: a possible link between inflammatory cytokines and atherogenesis. Proc Natl Acad Sci U S A 2000;97:8681-8686.
https://doi.org/10.1073/pnas.150098097

 

64 Deng X, Yin X, Allan R, Lu DD, Maurer CW, Haimovitz-Friedman A, Fuks Z, Shaham S, Kolesnick R: Ceramide biogenesis is required for radiation-induced apoptosis in the germ line of C. elegans. Science 2008;322:110-115.
https://doi.org/10.1126/science.1158111

 

65 Scheel-Toellner D, Wang K, Craddock R, Webb PR, McGettrick HM, Assi LK, Parkes N, Clough LE, Gulbins E, Salmon M, Lord JM: Reactive oxygen species limit neutrophil life span by activating death receptor signaling. Blood 2004;104:2557-2564.
https://doi.org/10.1182/blood-2004-01-0191

 

66 Schutze S, Potthoff K, Machleidt T, Berkovic D, Wiegmann K, Kronke M: TNF activates NF-kappa B by phosphatidylcholine-specific phospholipase C-induced "acidic" sphingomyelin breakdown. Cell 1992;71:765-776.
https://doi.org/10.1016/0092-8674(92)90553-O

 

67 Davis CN, Tabarean I, Gaidarova S, Behrens MM, Bartfai T: IL-1beta induces a MyD88-dependent and ceramide-mediated activation of Src in anterior hypothalamic neurons. J Neurochem 2006;98:1379-1389.
https://doi.org/10.1111/j.1471-4159.2006.03951.x

 

68 Grassme H, Jekle A, Riehle A, Schwarz H, Berger J, Sandhoff K, Kolesnick R, Gulbins E: CD95 signaling via ceramide-rich membrane rafts. J Biol Chem 2001;276:20589-20596.
https://doi.org/10.1074/jbc.M101207200

 

69 Grassme H, Jendrossek V, Bock J, Riehle A, Gulbins E: Ceramide-rich membrane rafts mediate CD40 clustering. J Immunol 2002;168:298-307.
https://doi.org/10.4049/jimmunol.168.1.298

 

70 Stancevic B, Kolesnick R: Ceramide-rich platforms in transmembrane signaling. FEBS Lett 2010;584:1728-1740.
https://doi.org/10.1016/j.febslet.2010.02.026

 

71 Zhang Y, Li X, Becker KA, Gulbins E: Ceramide-enriched membrane domains--structure and function. Biochim Biophys Acta 2009;1788:178-183.
https://doi.org/10.1016/j.bbamem.2008.07.030

 

72 Yu ZF, Nikolova-Karakashian M, Zhou D, Cheng G, Schuchman EH, Mattson MP: Pivotal role for acidic sphingomyelinase in cerebral ischemia-induced ceramide and cytokine production, and neuronal apoptosis. J Mol Neurosci 2000;15:85-97.
https://doi.org/10.1385/JMN:15:2:85

 

73 Adibhatla RM, Hatcher JF, Gusain A: Tricyclodecan-9-yl-xanthogenate (D609) mechanism of actions: a mini-review of literature. Neurochem Res 2012;37:671-679.
https://doi.org/10.1007/s11064-011-0659-z

 

74 Grassme H, Gulbins E, Brenner B, Ferlinz K, Sandhoff K, Harzer K, Lang F, Meyer TF: Acidic sphingomyelinase mediates entry of N. gonorrhoeae into nonphagocytic cells. Cell 1997;91:605-615.
https://doi.org/10.1016/S0092-8674(00)80448-1

 

75 Yu J, Novgorodov SA, Chudakova D, Zhu H, Bielawska A, Bielawski J, Obeid LM, Kindy MS, Gudz TI: JNK3 signaling pathway activates ceramide synthase leading to mitochondrial dysfunction. J Biol Chem 2007;282:25940-25949.
https://doi.org/10.1074/jbc.M701812200

 

76 Herr I, Martin-Villalba A, Kurz E, Roncaioli P, Schenkel J, Cifone MG, Debatin KM: FK506 prevents stroke-induced generation of ceramide and apoptosis signaling. Brain Res 1999;826:210-219.
https://doi.org/10.1016/S0006-8993(99)01288-3

 

77 Arii T, Kamiya T, Arii K, Ueda M, Nito C, Katsura KI, Katayama Y: Neuroprotective effect of immunosuppressant FK506 in transient focal ischemia in rat: therapeutic time window for FK506 in transient focal ischemia. Neurol Res 2001;23:755-760.
https://doi.org/10.1179/016164101101199135

 

78 Miyazawa N, Saji H, Takaishi Y, Nukui H: Protective effect of FK506 in the reperfusion model after short-term occlusion of middle cerebral artery in the rat: assessment by autoradiography using [125I]PK-11195. Neurol Res 2000;22:630-633.
https://doi.org/10.1080/01616412.2000.11740732

 

79 Nito C, Ueda M, Inaba T, Katsura K, Katayama Y: FK506 ameliorates oxidative damage and protects rat brain following transient focal cerebral ischemia. Neurol Res 2011;33:881-889.
https://doi.org/10.1179/1743132811Y.0000000019

 

80 Almawi WY, Melemedjian OK: Clinical and mechanistic differences between FK506 (tacrolimus) and cyclosporin A. Nephrol Dial Transplant 2000;15:1916-1918.
https://doi.org/10.1093/ndt/15.12.1916

 

81 Zawadzka M, Kaminska B: A novel mechanism of FK506-mediated neuroprotection: downregulation of cytokine expression in glial cells. Glia 2005;49:36-51.
https://doi.org/10.1002/glia.20092

 

82 Furuichi Y, Noto T, Li JY, Oku T, Ishiye M, Moriguchi A, Aramori I, Matsuoka N, Mutoh S, Yanagihara T: Multiple modes of action of tacrolimus (FK506) for neuroprotective action on ischemic damage after transient focal cerebral ischemia in rats. Brain Res 2004;1014:120-130.
https://doi.org/10.1016/j.brainres.2004.04.031

 

83 Tsujikawa A, Ogura Y, Hiroshiba N, Miyamoto K, Kiryu J, Honda Y: Tacrolimus (FK506) attenuates leukocyte accumulation after transient retinal ischemia. Stroke 1998;29:1431-1437; discussion 1437-1438.
https://doi.org/10.1161/01.STR.29.7.1431

 

84 Ohtani R, Tomimoto H, Kondo T, Wakita H, Akiguchi I, Shibasaki H, Okazaki T: Upregulation of ceramide and its regulating mechanism in a rat model of chronic cerebral ischemia. Brain Res 2004;1023:31-40.
https://doi.org/10.1016/j.brainres.2004.07.024

 

85 Nakane M, Kubota M, Nakagomi T, Tamura A, Hisaki H, Shimasaki H, Ueta N: Lethal forebrain ischemia stimulates sphingomyelin hydrolysis and ceramide generation in the gerbil hippocampus. Neurosci Lett 2000;296:89-92.
https://doi.org/10.1016/S0304-3940(00)01655-4

 

86 Lim CM, Kim SW, Park JY, Kim C, Yoon SH, Lee JK: Fluoxetine affords robust neuroprotection in the postischemic brain via its anti-inflammatory effect. J Neurosci Res 2009;87:1037-1045.
https://doi.org/10.1002/jnr.21899

 

87 Kim DH, Li H, Yoo KY, Lee BH, Hwang IK, Won MH: Effects of fluoxetine on ischemic cells and expressions in BDNF and some antioxidants in the gerbil hippocampal CA1 region induced by transient ischemia. Exp Neurol 2007;204:748-758.
https://doi.org/10.1016/j.expneurol.2007.01.008

 

88 Brunkhorst R, Friedlaender F, Ferreiros N, Schwalm S, Koch A, Grammatikos G, Toennes S, Foerch C, Pfeilschifter J, Pfeilschifter W: Alterations of the Ceramide Metabolism in the Peri-Infarct Cortex Are Independent of the Sphingomyelinase Pathway and Not Influenced by the Acid Sphingomyelinase Inhibitor Fluoxetine. Neural Plast 2015;2015:503079.
https://doi.org/10.1155/2015/503079

 

89 Windle V, Corbett D: Fluoxetine and recovery of motor function after focal ischemia in rats. Brain Res 2005;1044:25-32.
https://doi.org/10.1016/j.brainres.2005.02.060

 

90 Jolkkonen J, Puurunen K, Rantakomi S, Sirvio J, Haapalinna A, Sivenius J: Effects of fluoxetine on sensorimotor and spatial learning deficits following focal cerebral ischemia in rats. Restor Neurol Neurosci 2000;17:211-216.

 

91 Chollet F, Tardy J, Albucher JF, Thalamas C, Berard E, Lamy C, Bejot Y, Deltour S, Jaillard A, Niclot P, Guillon B, Moulin T, Marque P, Pariente J, Arnaud C, Loubinoux I: Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol 2011;10:123-130.
https://doi.org/10.1016/S1474-4422(10)70314-8

 

92 Pariente J, Loubinoux I, Carel C, Albucher JF, Leger A, Manelfe C, Rascol O, Chollet F: Fluoxetine modulates motor performance and cerebral activation of patients recovering from stroke. Ann Neurol 2001;50:718-729.
https://doi.org/10.1002/ana.1257

 

93 Jorge RE, Robinson RG, Arndt S, Starkstein S: Mortality and poststroke depression: a placebo-controlled trial of antidepressants. Am J Psychiatry 2003;160:1823-1829.
https://doi.org/10.1176/appi.ajp.160.10.1823

 

94 Leijon G, Boivie J: Central post-stroke pain--a controlled trial of amitriptyline and carbamazepine. Pain 1989;36:27-36.
https://doi.org/10.1016/0304-3959(89)90108-5

 

95 Focus Trial Collaboration: Effects of fluoxetine on functional outcomes after acute stroke (FOCUS): a pragmatic, double-blind, randomised, controlled trial. Lancet (London, England) 2019;393:265-274.

 

96 Robinson RG, Schultz SK, Castillo C, Kopel T, Kosier JT, Newman RM, Curdue K, Petracca G, Starkstein SE: Nortriptyline versus fluoxetine in the treatment of depression and in short-term recovery after stroke: a placebo-controlled, double-blind study. Am J Psychiatry 2000;157:351-359.
https://doi.org/10.1176/appi.ajp.157.3.351