Skip to main content
Erschienen in: Acta Neuropathologica 5/2019

Open Access 27.11.2018 | Review

Post-stroke inflammation—target or tool for therapy?

verfasst von: Kate Lykke Lambertsen, Bente Finsen, Bettina Hjelm Clausen

Erschienen in: Acta Neuropathologica | Ausgabe 5/2019

Abstract

Inflammation is currently considered a prime target for the development of new stroke therapies. In the acute phase of ischemic stroke, microglia are activated and then circulating immune cells invade the peri-infarct and infarct core. Resident and infiltrating cells together orchestrate the post-stroke inflammatory response, communicating with each other and the ischemic neurons, through soluble and membrane-bound signaling molecules, including cytokines. Inflammation can be both detrimental and beneficial at particular stages after a stroke. While it can contribute to expansion of the infarct, it is also responsible for infarct resolution, and influences remodeling and repair. Several pre-clinical and clinical proof-of-concept studies have suggested the effectiveness of pharmacological interventions that target inflammation post-stroke. Experimental evidence shows that targeting certain inflammatory cytokines, such as tumor necrosis factor, interleukin (IL)-1, IL-6, and IL-10, holds promise. However, as these cytokines possess non-redundant protective and immunoregulatory functions, their neutralization or augmentation carries a risk of unwanted side effects, and clinical translation is, therefore, challenging. This review summarizes the cell biology of the post-stroke inflammatory response and discusses pharmacological interventions targeting inflammation in the acute phase after a stroke that may be used alone or in combination with recanalization therapies. Development of next-generation immune therapies should ideally aim at selectively neutralizing pathogenic immune signaling, enhancing tissue preservation, promoting neurological recovery and leaving normal function intact.
Hinweise
Kate Lykke Lambertsen, Bente Finsen and Bettina Hjelm Clausen authors contributed equally.
This review is part of a review cluster on “Neuroinflammatory mechanisms in neurodegenerative disorders” edited by Dr. Roberta Brambilla.
Abkürzungen
Ab
Antibody
ATROSAB
Antagonistic TNF receptor one-specific antibody
BBB
Blood brain barrier
DWI
Diffusion-weighted imaging
gp130
Glycoprotein 130
icIL-1Ra
Intracellular interleukin-1 receptor antagonist
IL
Interleukin
IL-1R
Interleukin-1 receptor
IL-1Ra
Interleukin-1 receptor antagonist
IL-1RAcP
Interleukin-1 receptor accessory protein
IL-6R
Interleukin-6 receptor
IL-10R
Interleukin-10 receptor
i.c.v.
Intracerebroventricular
i.v.
Intravenous
ko
Knock out
LTα
Lymphotoxin-alpha
MCA
Middle cerebral artery
MCAO
Middle cerebral artery occlusion
NF-κB
Nuclear factor-kappa B
PET
Positron emission tomography
pMCAO
Permanent middle cerebral artery occlusion
PMN
Polymorphonuclear
PWI
Perfusion-weighted imaging
r
Recombinant
rh
Recombinant human
s.c.
Subcutaneous
sgp130
Soluble glycoprotein 130
sIL-1Ra
Secreted interleukin-1 receptor antagonist
sIL-6R
Soluble interleukin-6 receptor
solTNF
Soluble tumor necrosis factor
TACE
Tumor necrosis factor-alpha converting enzyme
TfR
Transferrin receptor
tmTNF
Transmembrane tumor necrosis factor
TNF
Tumor necrosis factor
TNFR
Tumor necrosis factor receptor
tMCAO
Transient middle cerebral artery occlusion
TROS
TNF receptor one silencer

Ischemic stroke

Ischemic stroke is the second leading cause of preventable deaths and the third leading cause of long-term disability worldwide [84]. This review focuses on the possibility of targeting post-stroke inflammation to improve tissue preservation, neurological outcome, and long-term survival. Ischemic stroke, accounting for approx. 90% of all stroke cases [84], is caused by embolism or thrombosis of a cerebral artery. This typically occurs in the middle cerebral artery (MCA), which supplies the lateral convexity of the cerebral hemisphere and thereby the majority of the primary motor and sensory cortex, leading to contralateral hemiplegia with reduced sensation. Today, recanalization by intravenous (i.v.) thrombolysis and thrombectomy are first-line treatments for ischemic stroke patients [95]. One of the major criteria for i.v. thrombolysis is the 4.5-h ‘therapeutic time window’, although the recent DAWN and DEFUSE 3 trials, which combine thrombectomy and i.v. thrombolysis, suggest expanding the therapeutic window up to 24 h when using perfusion imaging to guide treatment [2, 127]. Importantly, these studies additionally document that restoring perfusion not only leads to smaller infarcts, but that smaller infarcts correlate with a better neurological outcome [2, 127]. Given the low number of stroke patients eligible for treatment using thrombolysis and/or thrombectomy (approx. 10%), novel treatment options are critically needed. New therapies targeting key pathogenic mechanisms, including post-stroke inflammation, are currently being pursued experimentally and clinically, either alone or in combination with thrombolysis and/or thrombectomy [23]. Such treatments might also benefit stroke patients with good collateral blood supply who suffer permanent ischemia or patients in whom recanalization treatment is contraindicated.

The ischemic penumbra as target for post-stroke intervention

The ischemic penumbra consists of electrophysiologically silenced, potentially salvageable tissue [7], that can be assessed clinically using the “mismatch” between perfusion- and diffusion-weighted magnetic resonance images (PWI–DWI mismatch) [142] or positron emission tomography (PET) [77]. The cerebral metabolic rates for oxygen measured by PET define cerebral blood flow in cortical grey matter below 12 ml/100 g/min as infarct core, flow between 12 and 22 ml/10 g/min as critically hypoperfused penumbral tissue, and flow between 22 and 35 ml/100 g/min as an area of oligemia, i.e. hypoperfused tissue without risk for infarction [77]. This should be compared to the flow in normal grey matter, which is between 50 and 55 ml/100 g/min [95]. In tissue sections, the penumbra is defined as areas with reduced protein synthesis but preserved ATP content. This matches brain areas with transient heat shock protein 70 mRNA expression from 3 to 4 h after MCA occlusion (MCAO) [74, 75]. Using these definitions, the penumbra presents 30% of the final infarct volume at 1 h, approx. 18% at 6 h, and 5% at 24 h after permanent MCAO (pMCAO) [74]. After transient MCAO (tMCAO), the penumbra is initially increased as a result of edema associated with reperfusion, after which it is gradually recruited into the infarct and regresses to the final infarct volume at day 3 [75]. In rats, the infarct volume measured at 24 h after proximal pMCAO is significantly larger than after 60 min of proximal tMCAO, but is similar to that observed after 180 min of proximal tMCAO [112].
By showing that the therapeutic window can be expanded, the DAWN and DEFUSE 3 trial results, combining the use of thrombectomy and thrombolysis [2, 127], have ‘thrown the ball back in the ring’ in experimental stroke research. Some tMCAO models mirror thrombectomy in terms of reperfusion dynamics (review by [107]), encouraging testing of novel combination treatments. Furthermore, the clinical documentation that smaller infarcts translate into better neurological outcome [2, 127] emphasizes the importance of infarct volume reduction, ideally in conjunction with improved functional recovery, as an important outcome in experimental stroke research. The size of ischemic damage is typically presented as: 1—total infarct volume (‘direct infarct volume’ given in mm3), or 2—percentage of infarcted tissue in the ipsilateral hemisphere, corrected for edema formation and infarct resorption (‘indirect infarct volume’) (for details see [140]). Infarct volumes given as percentages and corrected for edema/resorption remain largely constant from 24 h to 24 weeks [140]. Direct infarct volumetric data obtained at 24 h after occlusion are robust, while data obtained at 5 days represents the cumulative effect of infarct formation and resorption [94, 140].

The inflammatory response in stroke

Inflammation is integral to the pathophysiology of ischemic stroke and a prime target for the development of new stroke therapies. The first immune cells to sense a stroke are the brain-resident microglial cells, which are innate immune cells that are perfectly situated and equipped to sense imbalances in the CNS. Microglia express receptors that are involved in immune signaling and modulation, recognition of danger signals elicited from dying cells, pathogens and self-antigens, as well as neurotransmitter receptors in both human [56] and mouse [78]. Like other cells, the microglia are sensitive to ischemia. 12 h after pMCAO, CD11b+ microglia in the infarct show signs of fragmentation, and by 24 h the number of microglia within the infarct is reduced [81, 94]. Microglia in the ‘peri-infarct’ show signs of activation in the form of process retraction from 30 min to 1 h after pMCAO, followed by upregulation of CD11b, CD45 and Iba1 in the peri-infarct from 3.5 to 6 h [32, 81, 94], where also the first CD11b+ macrophage-like cells (and Gr1+ neutrophils) appear [32, 94]. Microglial activation in the peri-infarct persists weeks after MCAO [94, 131]. Importantly, the microglia in the peri-infarct and infarct display different pro- and anti-inflammatory phenotypes [32, 33, 115], which include the expression of the pro-inflammatory cytokines tumor necrosis factor (TNF), interleukin (IL)-1β, and the anti-inflammatory IL-1 receptor antagonist (IL-1Ra) (Fig. 1) [32, 33, 92]. Microglia appear not to display classical M1 and M2 phenotypes after experimental stroke [61]. During later stages microglia, like monocytic macrophages, contribute to the resolution of the infarct by phagocytosing dead cells or debris, which is considered beneficial (review by [124]). However, microglia can also engulf viable ischemic neurons, that transiently express “eat-me” signals [122], and if dysregulated thereby increase neuronal cell death in the peri-infarct.
The infiltrating leukocytes, predominantly polymorphonuclear leukocytes (PMNs, neutrophils) and monocytes/macrophages, play different and complex roles in ischemic stroke. Neutrophils infiltrate early after MCAO [26]. They attach to the endothelium by binding different adhesion molecules (review by [125]), and with CXCL1 and CXCL2 as the main chemokines responsible for neutrophil extravasation [176]. Neutrophils expressing Ly6G and myeloperoxidase have been identified in the leptomeninges from 6 h after occlusion, thereafter in the Virchow–Robin spaces and superficial cortical layers, to eventually become widespread in the infarct and peri-infarct [133, 176]. In rodent pMCAO models, the number of neutrophils in infarct and peri-infarct peaks at 24 h and gradually decreases from 48 to 72 h [133, 176]. Differences in the peak of neutrophil recruitment have been reported between pMCAO and tMCAO [198]. Neutrophil accumulation has traditionally been considered detrimental post-stroke, either through the release of neurotoxic proteolytic enzymes [4] or neutrophil accumulation causing further blood flow obstruction and the ‘no-reflow’ phenomenon (reviewed in [39]). Neutrophils have also been shown to cause disruption of the blood–brain barrier (BBB) and hemorrhagic transformation post-stroke, worsening the neurological outcome [83]. Blockade of neutrophil recruitment has been shown to improve the functional outcome in rodent stroke models [83]. Neutropenia does not affect infarct size after MCAO [76] however, and none of the anti-neutrophil therapies tested so far have shown a beneficial effect in stroke patients [83]. Interestingly, neutrophils appear to display different phenotypes (neurotoxic N1 and neuroprotective N2) that may shape the effector functions of other cells and they are themselves cleared by phagocytic microglia/macrophages, which is considered important for the resolution of inflammation post-stroke [36]. Therefore, inhibiting neutrophil recruitment could also prove damaging if applied at the wrong time point.
Recruitment of circulating monocytes to the ischemic brain equals that of neutrophils and is regulated by adhesion molecules, chemokines, and cytokines. CD11b+Ly6ChighCCR2+ monocytes appear to be the predominant cell type recruited after both pMCAO and tMCAO [27, 116]. Recruitment after tMCAO takes place in a CCR2-dependent manner [41], while this appears not to be the case after pMCAO [27]. Histologically, CD11b+ and CD45+ macrophage-like cells are observed both in the infarct and peri-infarct from 6 to 48 h after pMCAO [94, 131]. From 3 to 7 days after occlusion the infarct becomes infiltrated with CD11b+, CD45+, and ED1+ macrophages, reminiscent of phagocytic ‘foam cells’ that are prominent in the infarct [81, 94]. Interestingly, when in the brain the Ly6ChighCCR2+ monocytes change their phenotype by downregulating Ly6C expression, upregulating F4/80, and then expressing arginase-1 and the chitinase-like protein YM-1, thereby developing into M2 phenotype macrophages [116]. This occurs from 1 to 3 days after pMCAO [116]. Histologically, Ym1+ and CD206+ cells have been shown to be abundant within the infarct core at 24 h, and to be even more numerous at 7 days, along with cells expressing the lysosomal marker CD68 [131]. This is in line with a role in infarct resolution and repair.
Although monocytes/macrophages have been reported to exacerbate ischemic brain damage in the acute phase after tMCAO [41], blocking the infiltration of Ly6Chigh monocytes (and neutrophils) using a CCR2 antagonist worsened the outcome after tMCAO, which was ascribed to CCR2 antagonism altering the polarization of infiltrated macrophages [27]. Monocytes/macrophages have been suggested to exert beneficial effects in the sub-acute phase after a stroke, by preventing hemorrhagic transformation [63], emphasizing that inhibition of monocyte recruitment might be damaging if done at the wrong time point. To add to the complexity, it appears that subsets of CD11b+CD45high macrophages express different pro- and anti-inflammatory cytokines at different time points after pMCAO [27, 32, 33, 92], raising the potential to modulate this expression and to stimulate the production of anti-inflammatory cytokines such as IL-1Ra [33]. The emerging understanding of how macrophages are stimulated by the ischemic environment to adopt distinct phenotypes or exert different functions might reveal new therapeutic strategies for controlling inflammation after ischemic injury.
Recent studies have also implicated lymphocytes in the pathogenesis of acute stroke. Since it is largely unknown as yet how these cells affect inflammation in the ischemic brain, the reader is referred to existing reviews on this topic [153].

Cytokines and cytokine therapies in experimental and human stroke

Treatment strategies aimed at preventing ischemia-induced cell death and promoting anti-inflammatory responses in ischemic tissue at risk have been studied both experimentally and in clinical trials (Table 1). Special attention has been given to inflammatory cytokines and the possibility to modulate their pro- or anti-inflammatory properties. Cytokine therapies are based on administration of highly specific engineered antibodies, soluble cytokine receptors, and mutant or fusion proteins that bind and neutralize the activities of a given cytokine (Table 2). A number of drugs targeting the key pro-inflammatory cytokines TNF, IL-1, and IL-6 (Table 2) are already being used in patients for the treatment of non-neurological diseases such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis. As cytokines have both beneficial and detrimental effects, their neutralization can result in unwanted side effects, including predisposing patients to infections, lupus-like syndrome, lymphoma, long-term effects on the cardiovascular system, and demyelinating disease [151]. Therefore, there is a need to develop and evaluate novel therapeutics that can better distinguish between detrimental and protective signaling of a given cytokine. Four cytokines have proven especially promising as potential therapeutic targets in experimental ischemic stroke: the pro-inflammatory cytokines TNF, IL-1, IL-6 and the anti-inflammatory cytokine IL-10.
Table 1
Studies on anti-cytokine treatments in experimental and human stroke
Ischemia model
Strain
Intervention
Results
Target involved
References
TNF system
 Mouse
  Distal pMCAO (electrocoagulation)
C57BL/6
i.v. injection of 10 mg/kg anti-TNF inhibitor (etanercept) or 10 mg/kg anti-solTNF inhibitor (XPro1595) 30 min after occlusion
No change in infarct volume, improved functional outcome
tmTNF and/or solTNF
[30]
  Proximal tMCAO (60 min, filament)
C57BL/6
i.v. injection of 1 mg/kg etanercept or cTfRMab-TNFR 45 or 90 min after occlusion
cTfRMAb-TNFR decreased infarct volume and neural deficits
tmTNF and solTNF
[167]
  Proximal tMCAO (60 min, filament)
C57BL/6
i.v. injection of 1 mg/kg cTfRMab-TNFR and 1 mg/kg cTfRMab-GDNF 45 min after occlusion
cTfRMAb-TNFR and cTfRMAb-GDNF decreased infarct volume
mTNF and solTNF
[168]
  Cortical photothrombosis (i.v. Bengal Rose injection followed by 20 min focal illumination)
C57BL/6
Intracortical infusion of 1 μg/day sTNF-α- R1 for 1 week
sTNF-α- R1 preserved post-stroke deprivation-induced brain plasticity
solTNF (and tmTNF)
[98]
  Distal pMCAO (electrocoagulation)
BALB/c
i.p. or i.v. injection of 3 mg/kg TNF-bp immediately after occlusion
TNF-bp decreased infarct volume
tmTNF and solTNF
[120]
  Distal pMCAO (electrocoagulation)
BALB/c
Topic administration of 3 mg/kg TNF-bp immediately and 1 h after occlusion
TNF-bp decreased infarct volume
tmTNF and solTNF
[121]
 Rat
  Proximal tMCAO (90 min, filament)
Wistar
i.p. injection of 7 mg/kg chimeric anti-TNF mAb (infliximab) or 5 mg/kg anti-TNF (etanercept) 0 and 6 hrs after occlusion
Infliximab and etanercept decreased infarct volume
tmTNF and solTNF
[5]
  Proximal tMCAO (120 min, filament)
SD (diabetic and non-diabetic)
i.p. or i.v. injection of 300, 450, or 900 μg/kg anti-TNF (etanercept) within 24 hrs before or immediately after occlusion
Etanercept administered once before occlusion reduced infarct volume in non-diabetic rats and at 900 μg/kg/daily for 5 weeks prior to occlusion decreased infarct volume in diabetic rats
tmTNF and solTNF
[82]
   Distal tMCAO (occluded and cut)
SHR
10 μg TNF mAb or 12.5 μg solTNFR1, 30 min before and 3 and 6 h after occlusion
TNF mAb and solTNFR1 decreased infarct volumes
tmTNF and solTNF
[8]
   Proximal tMCAO (60 min, filament)
SD
i.v. injection of ex vivo-derived dendritic cells (exDCs) overexpressing solTNFR1 6 h after reperfusion
solTNFR1-exDCs decreased infarct size and inflammation
solTNF and (tmTNF)
[186]
   Proximal tMCAO (120 min, filament)
SD
i.v. injection of 15 mg/kg anti-TNF mAb immediately after reperfusion
Anti-TNF mAb decreased infarct volume and edema
tmTNF and solTNF
[79]
 Human
  Chronic stroke (13-36 months old)
 
Perispinal, interspinous, extrathecal injection of 25 mg anti-TNF (etanercept)
Neurological improvement in all patients (n = 3)
tmTNF and solTNF
[173]
  Chronic stroke (≤3 to >120 months)
 
Perispinal, interspinous, extrathecal injection of 25 mg anti-TNF (etanercept)
Improved motor impairment, spasticity, sensory impairment, cognition, psychological/behavioral function, aphasia, and pain (n=617)
tmTNF and solTNF
[174]
IL-1 system
 Mouse
  Distal tMCAO (30 and 45 min, filament)
C57BL/6
s.c. injection of 100 mg/kg IL-1Ra 30 or 180 min after
IL-1Ra decreased infarct size and neurological deficit and improved functional outcome
IL-1α, IL-1β
[106]
  Distal pMCAO (electrocoagulation)
BALB/c
s.c. injection of 100 mg/kg IL-1Ra 30 or 180 min after
  Distal pMCAO (electrocoagulation)
C57BL/6
i.v. injection of IL-1Ra-producing bone marrow-derived cells 30 min after occlusion
IL-1Ra-producing bone marrow-derived cells decreased infarct volumes and improved functional outcomes
IL-1α, IL-1β
[33]
   Proximal tMCAO (40 min, filament)
C57BL/6
i.v. injection of IL-1Ra-producing bone marrow-derived cells 30 min after reperfusion
  Proximal tMCAO (30 min, filament)
C57xSV129
i.c.v. injection of 2.5 μg IL-1Ra or 2.5 ng IL-1β 30 min before occlusion and 10 min after reperfusion
IL-1β increased, whereas IL-1Ra decreased infarct volumes
IL-1α, IL-1β
[175]
 Rats
  Proximal tMCAO (120 min, filament)
SD
i.v. injection of 50 mg/kg IL-1RA-PEP at the time of reperfusion
IL-1RA-PEP alleviated brain infarction, cerebral edema, neurological deficit score, and motor performance
IL-1β
[195]
  Proximal tMCAO (filament)
SD
i.v. injection of 10 mg at the time of occlusion followed by i.v. infusion 0.8 mg/h hIL-1Ra (anakinra) for 24 hrs
Anakinra reduced infarct volume
IL-1α, IL-1β
[28]
  Proximal tMCAO (120 min, filament)
Wistar
i.v. injection of 5, 10, or 20 mg/kg hIL-1Ra (anakinra) at 3, 6 or 12 hrs after after occlusion
Anakinra reduced infarct volume and improved neurological deficits dose- and time-dependently
IL-1α, IL-1β
[189]
  Proximal tMCAO (120 min, filament)
SD
i.v. injection of 50 mg/kg IL-1RA-PEP at the time of reperfusion
IL-1RA-PEP alleviated brain infarction, cerebral edema, neurological deficit score and motor performance
IL-1α, IL-1β
[195]
  Distal pMCAO (electrocoagulation)
SD
i.c.v. injection of 10 μg rhIL-1Ra 30 min before and 10 min after occlusion
rhIL-1Ra reduced infarct volumes
IL-1α, IL-1β
[138]
   Distal tMCAO (60 min, filament)
SD
i.c.v. injection of recombinant adenovirus vector carrying the human IL-1Ra cDNA (Ad.RSVIL-1ra) 5 days prior to experimental stroke
Ad.RSVIL-1ra reduced infarct volumes
IL-1α, IL-1β
[12]
  Proximal pMCAO (filament)
Wistar
i.v. injection of 100 mg/kg rhIL-1Ra immediately prior to and again s.c. 3 times per day for 7 days
rhIL-1Ra reduced infarct volumes and improved functional scores
IL-1α, IL-1β
[59]
  Distal pMCAO (electrocoagulation)
SD
s.c. injection of 100 mg/kg rhIL-1Ra at 0, 4, 8, 12, and 18 h after occlusion
rhIL-1Ra reduced infarct volumes dose- and time- dependently and inhibited cerebral edema at 24 hrs
IL-1α, IL-1β
[137]
 Human
  Acute stroke (< 6 h)
 
i.v. injection of 100 mg bolus rhIL-1Ra, followed by 2 mg/kg per hour for 72 h
rhIL-1Ra improved clinical outcomes (survival to 3 months, NIHSS, BI, and mRS scores) at 3 months (n=17)
IL-1α, IL-1β
[51]
  Acute stroke (< 6 h)
 
i.v. injection of 100 mg bolus rhIL-1Ra, followed by 2 mg/kg per hour for 72 h
rhIL-1Ra reversed peripheral innate immune suppression in the acute phase of stroke (n=17)
IL-1α, IL-1β
[158]
  Acute stroke (< 5 h)
 
s.c. injection of 100 mg rhIL-1Ra (anakinra) twice daily for 3 days
Anakinra reduced plasma inflammatory markers but did not affect mRS at 3 months (n=39)
IL-1α, IL-1β
[159]
IL-6 system
 Mouse
   Distal pMCAO (electrocoagulation)
C57BL/6
i.v. injection of 500 ng IL-6, solIL-6R, or 500 ng IL-6 followed by 500 ng solIL-6R 5 min or 5 and 60 min after occlusion
IL-6 injection improved behavioral outcome without affecting infarct size; co-administration of Il-6 and solIL-6R increased infarct volume, number of PMNs and impaired endurance
IL-6, IL-6R, gp130
[70]
  Proximal tMCAO (60 min, filament)
C57BL/6
i.c.v. injection of 10 ng anti-IL6 mAb or intranasal administration of 0.1 μg rIL-6 every 24 h for 2 weeks starting 14 days after occlusion
Anti-IL-6 mAb reduced proliferation and neuronal differentiation of neural progenitor cells in the ipsilateral SVZ, as well as functional recovery; rIL-6 conferred the opposite effect
IL-6
[111]
 Proximal tMCAO (45 min, filament)
C57BL/6
i.p. injection of 100 μg/g bodyweight IL-6Ra immediately after reperfusion
Anti-IL-6Ra increased infarct volume and affected neurological function.
IL-6R
[192]
 Rats
  Proximal tMCAO (120 min, filament)
SD
i.p. injection of 50 or 500 ng rIL-6
rIL-6 reduced infarct volumes
IL-6R
[53]
  Proximal pMCAO (electrocoagulation)
SD
i.c.v. injection of 2x50 or 2x500 ng rhIL-6 30 min prior to and again 15 min after occlusion
rhIL-6 reduced infarct volumes
IL-6R
[100]
IL-10 system
 Mouse
  Distal pMCAO (electrocoagulation)
C57BL/6
i.c.v. injection of 100 ng rmIL-10 5 min after occlusion
rmIL-10 reduced infarct volumes
IL-10R
[96]
  Proximal tMCAO (60 min, filament)
C57BL/6
i.v. infection of IL-10-producing B cells 24 h prior to or 4 h after occlusion
IL-10-producing B cells reduced infarct volumes and reduced post-stroke inflammation
IL-10R
[16]
 Rats
     
  Distal tMCAO (90 min, filament)
SD
i.v. injection of IL-10-overproducing mesenchymal stem cells 0 or 3 h after reperfusion
IL-10-overproducing mesenchymal stem cells reduced infarct volumes, improved motor functions and reduced inflammation
IL-10R
[119]
  Distal pMCAO (photothrombotic)
SHR
i.c.v. injection of adenoviral vectors encoding human IL-10 (AdlIL-10) 90 min after occlusion
AdlL10 reduced infarct volumes and leukocyte infiltration
IL-10R
[130]
  Distal pMCAO (electrocoagulation)
SHR
i.c.v. injection of 1 μg IL-10 30 min and 3 hours after occlusion and i.v. injection of 5 or 15 μg/h for 3 h starting 30 min after occlusion
IL-10 treatments reduced infarct volumes
IL-10R
[160]
Ab antibody, BI Barthel index, bp binding protein, cTfRMAb transferrin receptor monoclonal antibody, GDNF glial-derived neurotropic factor, h human, i.c.v intracerebroventricular, IL interleukin, IL-1Ra interleukin-1 receptor antagonist, IL-1RI interleukin-1 receptor 1, IL-6R interleukin-6 receptor, IL-10R interleukin-10 receptor, i.p. intraperitoneal, i.v. intravenous, mAb monoclonal antibody, mRS modified rankin score, NIHSS National Institutes of Health Stroke Scale, pMCAO permanent middle cerebral artery occlusion, rh recombinant human, rm recombinant mouse, s.c. subcutaneous, SD Sprague–Dawley, SHR spontaneously hypertensive rats, solTNF soluble tumor necrosis factor, SVZ subventricular zone, tMCAO transient middle cerebral artery occlusion, tmTNF transmembrane tumor necrosis factor, TNF tumor necrosis factor, TNFR tumor necrosis factor receptor
Table 2
Mechanistic profile of cytokine and cytokine receptor agonists/antagonists for use in experimental stroke
Drug name
Class
Structure
Specificity
References
Etanercepta and biosimilars
Dimeric Fc-fusion protein
Hu TNFR2exc:IgG1-Fcγ1
solTNF, tmTNF, LTα3, & LTα2β1
 
Infliximaba and biosimilars
Monoclonal antibody
Mo/Hu chimeric IgG1/κ
solTNF & tmTNF
 
Adalimumaba and biosimilars
Monoclonal antibody
Hu IgG1/κ
solTNF & tmTNF
 
Certolizumab pegola
Monoclonal antibody fragment
PEGylated hu IgG1/κ Fab´
solTNF & tmTNF
 
Golimumaba
Monoclonal antibody
Hu IgG1/κ
solTNF & (tmTNF)
 
XPro1595
Dominant-negative inhibitor
TNF mutein
solTNF
[162]
XEN345
Dominant-negative inhibitor
TNF mutein
solTNF
[162]
cTfRMAb-TNFR
Fusion cTfR-protein
TNFR2exc:IgG1-cTfR
solTNF & tmTNF
[197]
R1antTNF
Inhibitor
TNFR1 selective mutein
TNFR1, solTNF?
[155]
DMS5540
Monovalent domain antibody
TNFR1-dAb:Albu-dAb
TNFR1
[108]
TROS
Dimeric nanobody
Hu TNFR1-Nb:Alb-70-96-Nb IgG1
TNFR1
[163]
ATROSAB
Monoclonal antibody
Hu IgG1
TNFR1
[88]
EHD2-scTNFR2
Dimeric single-chain fusion protein
Hu TNFR2:EHD2 IgE
TNFR2
[44]
TNCscTNF80
Trimerized single-chain fusion protein
Chicken TNC:huTNFR2
TNFR2
[25]
Anakinraa
Recombinant protein
IL-1Ra mutein
IL-1R1
 
Rilonacepta
Dimeric fusion protein
Hu IL-1R1excIL-1RAcPexc:IgG1-Fc
IL-1α & IL-1β
 
Canakinumaba
Monoclonal antibody
Hu IgG1/κ
IL-1β
 
MEDI-8968
Monoclonal antibody
Hu IgG2
IL-1R1
[21]
Gevokizumab
Monoclonal antibody
Hu IgG2/κ
IL-1β
[144]
LY2189102
Monoclonal antibody
Hu IgG4
IL-1β
[156]
XOMA 052
Monoclonal antibody
Hu IgG2/κ
IL-1β
[144]
IL-1RA-PEP
Fusion protein
IL-1Ra:PEP-1
IL-1R1
[195]
Tocilizumaba
Monoclonal antibody
Hu IgG1/κ
tmIL-6R & solIL-6R
 
Siltuximaba
Monoclonal antibody
Mo/Hu chimeric IgG1/κ
IL-6
 
Sarilumaba
Monoclonal antibody
Hu IgG1/κ
IL-6R
 
Olokizumab
Monoclonal antibody
Hu IgG1/κ
IL-6, gp130
[154]
Elsilimomab
Monoclonal antibody
Hu IgG1/κ
IL-6
[184]
Sirukumab
Monoclonal antibody
Hu IgG1/κ
solIL-6
[190]
Clazakizumab
Monoclonal antibody
Hu IgG1/κ
IL-6
[110]
sgp130Fc (Olamkicept)
Fusion protein
Hu gp130exc:IgG1-Fc
IL-6/solIL-6R complex
[86]
Pegliodecakin (AM0010)
Pegylated recombinant protein
PEG-rHuIL-10
IL-10R
[118]
PEGylated-IL10
Pegylated recombinant protein
PEG-rMuIL-10
IL-10R
[50]
Albu anti-serum albumin, cTfR transferrin receptor, dAb domain antibody, gp130 glycoprotein 130, Hu human, IL Interleukin, IL-1R interleukin-1 receptor, IL-1Ra interleukin-1 receptor antagonist, IL-1RAcP IL-1 receptor accessory protein, LTα lymphotoxin-alpha, Mo mouse, solIL-6R soluble interleukin-6 receptor, solTNF soluble tumor necrosis factor, tmIL-6R transmembrane interleukin-6 receptor, tmTNF transmembrane tumor necrosis factor, TNC tenascin, TNF tumor necrosis factor, TNFR tumor necrosis factor receptor
aFDA approved drug

Tumor necrosis factor

The most extensively studied cytokine in experimental stroke is the proinflammatory and immune regulatory cytokine TNF. It exists in a secreted form (solTNF) and a transmembrane form (tmTNF), which is also involved in reverse signaling [87]. solTNF is derived from tmTNF, which is cleaved by the protease ADAM-17, also known as TNF-alpha converting enzyme (TACE) [14]. tmTNF and solTNF signals are transmitted through two distinct receptors, TNFR1 and TNFR2, that differ significantly both in cellular expression and downstream effects. Although solTNF binds both receptors with high affinity, it preferentially binds to TNFR1 (dissociation constant [Kd] 20 pM) versus TNFR2 ([Kd] ~ 400 pM), owing to a 30-fold faster dissociation rate from TNFR2 than from TNFR1 [69]. This has given rise to a ligand-passing hypothesis, stating that solTNF binding to TNFR2 is quickly passed to TNFR1. Binding by TNFRs to tmTNF or even TNF antagonists can induce reverse signaling through tmTNF, leading to cell activation, cytokine suppression, or apoptosis of the tmTNF-bearing cell (reviewed in [49]). While TNFR1 is expressed on virtually all cells, TNFR2 expression is restricted to cells of the immune system, glial cells, and endothelial cells. TNF’s proinflammatory effects are likely mediated through solTNF–TNFR1 signaling, leading to activation of two major, well-understood pathways. One leads to the induction of anti-apoptotic genes, mainly through activation of the transcription factor nuclear factor-kappa B (NF-κB). The second signaling pathway results in activation of cellular suicide programs, including the prototype of programmed cell death, apoptosis, but also the execution of programmed necrosis (necroptosis) [179]. Under physiological conditions, TNF does not induce cell death unless transcription, translation, or specifically the NF-κB pathway are blocked. Unlike TNFR1, TNFR2 is not associated with induction of apoptosis but preferentially promotes cell growth, and regeneration through NF-κB activation. TNFR1 can be activated by binding of either solTNF or tmTNF, whereas TNFR2 is only fully activated by tmTNF [68, 69]. A further level of complexity is added by the proteolytic cleavage of the extracellular domains of both TNFR1 and TNFR2 [182], which is increased upon TNFR activation (reviewed by [1]). The soluble TNFR1 and solTNFR2 ectodomains that are shedded can bind to TNF, albeit with low affinity, and can thus act as natural inhibitors of TNF.
Lymphotoxin-alpha (LTα), another TNFR agonist with important roles in immune regulation, also binds TNFR1 and TNFR2 and mainly mediates NF-κB-mediated signaling [134].

Tumor necrosis factor in experimental stroke

The low baseline levels of TNF in the CNS under physiological conditions play an important role in neuronal function, by modulating glutamatergic synaptic transmission and plasticity [164]. Furthermore, TNF regulates neuronal networks involved in cognition and behavior [9], thereby attributing importance to TNF both in the healthy and stroke-lesioned CNS. Multiple checks are in place to finetune TNF’s production and activity, including regulation of TNF gene expression at transcriptional and translational levels, and the regulated shedding of TNF [117] and its receptors [135].
A particular role of TNF/TNFR1 in the etiopathogenesis of stroke is suggested by genome-wide association studies that found a polymorphism in the TNF gene that increases the susceptibility for stroke [178]. After pMCAO, TNF is acutely and significantly upregulated, peaks at 12–24 h (Fig. 2a), and remains elevated for days (Fig. 1d), making TNF a key player both in acute and chronic ischemia and in post-ischemic neuronal plasticity (reviewed by [91]). TNF is primarily produced by microglia in the early phase after experimental stroke and sustained by macrophages at later time points [20, 32, 92, 94], although other cell types like ependymal, astroglial and neuronal cells have also been reported to produce TNF during ischemic conditions (reviewed by [91]).
The use of genetically modified mice has been invaluable for establishing the role of TNF in the pathogenesis of ischemic stroke. Conventional TNF-knock out (KO) mice [92] and conditional TNF-KO mice with ablation of TNF in myeloid cells, including microglia [31] have larger infarcts and worse behavioral deficits than control mice after pMCAO. This suggests a neuroprotective role of microglial-derived TNF in ischemic stroke, an effect which appears to be mediated via TNFR1 [92, 170]. Interestingly, mice with a loss of TACE-mediated cleavage preventing shedding of solTNF (and thus expressing only tmTNF) develop smaller infarcts than their littermates [104], suggesting that removal of solTNF but preservation of tmTNF is neuroprotective in ischemic stroke.
Finally, a polymorphism in the LTα gene (LTA) has been linked to increased susceptibility for stroke [178], suggesting that also LTα plays a role in the etiopathogenesis of stroke. However, LTα levels appear to remain relatively constant in the acute phase after pMCAO in mice (Fig. 2a, Lambertsen et al., unpublished data), suggesting that brain-derived LTα has no major role in the inflammatory response post-stroke.

Anti-tumor necrosis factor treatment in experimental stroke

The currently used FDA- and EMA-approved anti-TNF therapeutics block both solTNF and tmTNF (Table 2). These therapeutics appear to relieve fatigue and symptoms of depression that can be associated with chronic inflammatory diseases [177]. Despite reports of improved neurological outcome in patients with stroke or traumatic brain injury who are treated with perispinal etanercept [172, 174] (Table 1), none of the currently used anti-TNF therapeutics have so far been approved as a neuroprotective strategy in combination with tissue plasminogen activator treatment. This may be because targeting both solTNF and tmTNF can predispose patients to both cardiovascular and demyelinating diseases [151], which is in line with the finding that a single nucleotide polymorphism in the TNFR1 gene (TNFRSF1A) that mimics the effect of anti-TNF therapeutics, is a risk factor for developing multiple sclerosis [67]. In combination with the observation that not only TNF-KO mice but also TNF-R1 KO mice develop larger infarcts than wild-type mice [92, 170], this calls for precaution in using the currently approved anti-TNF therapeutics and emphasizes the need for more specific anti-TNF therapeutics.
There has been little preclinical testing of therapeutics that exclusively target solTNF (XPro1595, XEN345, and possibly R1antTNF) (Tables 1, 2 and Fig. 3a) and leave signaling via tmTNF–TNFR1/2 intact. A comparative study of a single i.v. dose of XPro1595 (a dominant-negative solTNF inhibitor) or etanercept, administered at a dose of 10 mg/kg, 30 min after pMCAO, showed that both compounds affected the inflammatory response and improved motor functions and motor learning skills compared to vehicle 1 and 5 days after pMCAO, but had no effect on infarct volume [30]. This indicates that targeting solTNF alone may be efficient for the treatment of post-stroke inflammation. Similarly, recent findings showed that topical, but not systemic administration, of XPro1595 can rescue tissue at risk after experimental spinal cord injury, while etanercept had no effect [129], suggesting that topical administration of XPro1595 can inhibit solTNF present locally in the CNS. Clearly, more studies are needed to clarify whether XPro1595 is able to rescue tissue at risk in the peri-infarct. However, given the prevalence of post-stroke infections in humans, leaving tmTNF signaling intact may decrease the risk of infections.
While it seems relevant to retain the neuroprotective TNFR1 signaling in the acute phase after stroke, TNFR1 also plays a role in sustaining chronic inflammation in mouse models of multiple sclerosis and TNFR2 is important for remyelination [18]. Although more studies are clearly required to clarify the role of neuronal TNFR1 signaling in the acute phase post-stroke, it is possible that TNFR1-specific antagonists [R1antTNF, DMS5540, TROS (TNF receptor one silencer), ATROSAB (antagonistic TNF receptor one-specific antibody)] (Table 2) that preserve TNFR2 signaling, will be important in improving neuronal and synaptic remodeling in the chronic phase of stroke.
Due to their large size, many biologic TNF inhibitors do not cross the BBB and must be modified to enable BBB penetration and access to the brain parenchyma. One such drug is cTfRMAb-TNFR (Table 2), which ferries TNFR across the BBB using the transferrin receptor (TfR) [197]. In a preclinical study, i.v. injection of cTfRMAb-TNFR was compared to etanercept in a tMCAO model and when administered 90 min after occlusion resulted in reduced infarct volumes and reduced neural deficit 1 and 7 days post-stroke, whereas etanercept had no effect [167](Table 1). Despite the fact that both cTfRMAb-TNFR and etanercept are TNFR2 fusion proteins, the authors ascribed the beneficial effect of cTfRMAb-TNFR to the modification of this protein to allow it to be transported across the BBB [15].
In another preclinical study, sTNF-α R1 (solTNFR1) (Table 2) administered by intracortical infusion for 1 week after photothrombotic stroke was found to preserve deprivation-induced axonal plasticity in the cerebral cortex post-stroke [98] (Table 1). This effect was ascribed to sTNF-α R1 competing for solTNF with TNFR1 receptors, supporting the hypothesis that ablating solTNF is beneficial in ischemic stroke. This is in line with a preclinical study showing that intra-arterial injection of solTNFR1-overexpressing dendritic cells 6 h after tMCAO reduces infarct size and inflammation 3 days post-stroke [186] (Table 1).

Interleukin-1

The IL-1 family comprises 11 members (IL-1α, IL-1β, IL-1 receptor antagonist (IL-1Ra), IL-18, IL-33, IL-36α, IL-36β, IL-36γ, IL36-Ra, IL-37, and IL-38), forming a network of proinflammatory cytokines that regulate innate immune cells and function as key players in inflammation (review by [43]). Despite structural and functional similarities and evidence of a common ancestry [143], so far only IL-1α, IL-1β, and IL-1Ra have been studied extensively in ischemic stroke.
Both IL-1α and IL-1β are expressed and translated as precursor (pro) proteins. ProIL-1α is biologically active, but it lacks the signal peptide that allows it to leave the cell [143]. IL-1α is a ‘dual-function’ cytokine with both nuclear and cytoplasmic functions, but danger signals from necrotic cells can promote the secretion of IL-1α [48], causing neutrophil recruitment and exacerbation of inflammation [24]. Apoptosis causes IL-1α to translocate to the nucleus, where it binds to chromatin, a mechanism which is known to restrain inflammation [34]. IL-1α is considered to be an early danger signal that modulates a wide range of inflammatory reactions through the interleukin-1 receptor type 1 (IL-1R1) [48, 143]. Following injury, the proteolytic cleavage of IL-1α occurs through the actions of calpain, and possibly inflammasomes [194]. Membrane-bound, unprocessed IL-1α acts in a paracrine fashion on IL-1R expressing cells [42] to modulate angiogenesis, cell proliferation, senescence, apoptosis, and migration, and cytokine production ([149] and review by [43]).
In contrast to proIL-1α, proIL-1β is a biologically inactive protein, and both proIL-1β and mature IL-1β appear extracellularly [143], indicating that processing can take place after secretion. ProIL-1β is cleaved by caspase-1 (or IL-1 converting enzyme) [143], which gets activated by the assembly of the inflammasome, a process triggered in turn by damage-associated molecular pattern signals [72]. ProIL-1β can also be cleaved by neutrophil serine proteases such as proteinase 3 and elastase [123].
The natural regulator of IL-1 is IL-1Ra, which is found in two structural variants, secreted (s)IL-1Ra and intracellular IL-1Ra (icIL-1Ra), that both target the IL-1R1 [6]. The icIL-1Ra isoform is less explored but believed to exert multiple functions inside the cell [6], such as modulating the effect of IL-1α and/or acting as regulator of proIL-1β [102]. IL-1Ra is expressed by monocytes/macrophages, neutrophils [105], microglia [33], and other cells [42].
IL-1α/β induce their biological effects through IL-1R1, which is expressed in low numbers (< 100) on nearly all cells in the brain [42]. Binding of IL-1 to IL-1R1 allows the binding of the interleukin-1 receptor accessory protein (IL-1RAcP, IL-1R3), which is a key component of the receptor/agonist signaling complex [6, 143]. Recruitment and binding of IL-1RAcP converts the low-affinity binding between IL-1R1 and IL-1 to a high-affinity binding allowing further signal transduction [65]. IL-1 signaling is complex but potent with < 10 receptors/cell required to be occupied before a full response is triggered [166]. This means that IL-1Ra needs to be present in 100–1,000-fold molar excess to control its biological properties [42].
IL-1R2 shares structural characteristics with IL-1R1, but it lacks the cytoplasmic domain that allows signal transduction. IL-1R2 binds IL-1 as a decoy receptor [42, 143]. IL-1R2 is expressed by the same cells as IL-1R1 but is particularly abundant on monocytes, and neutrophils [42, 45]. IL-1R2 binds IL-1α in the cytosol, preventing its interaction with IL-1R1 when released from necrotic cells [196]. All the IL-1Rs are also found in a soluble form [90].

Interleukin-1 in experimental stroke

IL-1 is a major player in stroke pathology (Fig. 1g, h). As for the TNF gene, a polymorphism in the IL-1A gene has been associated with an increased susceptibility for ischemic stroke [199] whereas a polymorphism in the IL-1B gene has been associated with lower stroke risk [13], although this is still controversial [193, 199]. Polymorphisms in the IL1RN gene do not affect the risk for stroke [199], but increased plasma IL-1α combined with a polymorphism in the IL1RN gene increases the risk of post-stroke infection [10].
So far, focus has been on understanding the role of IL-1β in experimental stroke models, however data suggests that also IL-1α, which is significantly upregulated in mice 6–24 h after pMCAO (Fig. 2b) [33] and 7 days after tMCAO [149], plays an important role in stroke-induced neuroinflammation [33, 171]. Following experimental stroke in rodents, IL-1α was shown to be expressed by platelets and microglia [33, 40]. The presence of platelet-derived IL-1α acutely (6 h) after experimental stroke [33] supports findings that IL-1α drives neurovascular inflammation and facilitates neutrophil infiltration into the ischemic brain [171]. At 24 h after pMCAO, microglia are the key producers of IL-1, with approximately 50% of the IL-1α producing microglia co-expressing IL-1Ra and 17% co-expressing IL-1β, demonstrating that IL-1β and IL-1α are largely produced by segregated populations of microglia in the ischemic brain [33]. It is, therefore, likely that IL-1α in platelets in addition to few IL-1α/β producing microglia impacts the balance between IL-1/IL-1Ra early after stroke onset [33]. Findings that IL-1α and IL-1Ra are co-expressed in microglia support the view that icIL-1Ra can regulate the action of intracellular IL-1α [113].
IL-1β is constitutively expressed in the CNS [42] where it exerts neurotrophic factor-like activity [161] or regulates both the expression and activity of ion channels [181]. IL-1β is upregulated acutely after ischemic stroke (Fig. 1)[32, 33, 37] and peaks at 12-24 h (Fig. 2b) primarily in microglia and macrophages [32, 37], and later in astroglial-like cells [183].
IL-1 has been shown to aggravate stroke pathology (Table 1) as demonstrated by findings in transgenic mice overexpressing a dominant-negative form of caspase-1 in neurons [54], caspase-1 KO mice [73], and IL-1α/β KO mice [17], which all show reduced infarct volumes after experimental stroke. Additional support comes from early studies demonstrating that administration of recombinant IL-1β exacerbated damage [99] as does intracerebroventricular (i.c.v.) delivery of an IL-1Ra antiserum [101]. Systemic administration of IL-1β just before tMCAO worsened outcome in rodents through neutrophil- and platelet-dependent mechanisms reducing reperfusion [109].
In addition, IL-1Ra is an acute phase protein [55] that blocks the action of IL-1. Administration of IL-1Ra reduced ischemic brain damage after both tMCAO and pMCAO in rats [59, 137] and mice [175] (Table 1) and IL-1Ra-overexpressing mice show reduced infarct volumes, whereas IL-1Ra KO mice display increased infarct volumes compared to littermate mice after pMCAO [33].

Anti-interleukin-1 treatment in experimental and human ischemic stroke

IL-1Ra is the only therapeutic agent directed against IL-1-induced inflammation (Fig. 3b) that has been tested in randomized clinical trials in ischemic stroke (Table 1). In pre-clinical stroke models, recombinant (r)IL-1Ra is protective after central [137] and peripheral [59] administration and, similar to i.c.v. injection of anti-IL-1β antibody (Ab) [191] or IL-1Ra, was shown to reduce infarct volumes after MCAO in rats [99, 137] and pMCAO in mice [121].
Although IL-1Ra can reach the brain after systemic administration in the rat [66] and modulates long-term functional recovery after experimental stroke [62], its use in stroke patients has proven challenging. Pharmacokinetic studies have shown that rIL-1Ra crosses the BBB slowly [71] and has a very short half-life in the circulation [64], and thus it is difficult to achieve therapeutic IL-1Ra concentrations in the brain [57].
The first randomized, double-blind, placebo-controlled trial using i.v. injected recombinant human (rh)IL-1Ra in acute stroke patients (given within the first 6 h of stroke onset) showed a reduction in neutrophil count, plasma CRP, and IL-6 compared to placebo, and exploratory efficacy analysis indicated that patients receiving rhIL-1Ra had minimal to no disability three months after stroke [51]. Recently, the SCIL-STROKE (subcutaneous interleukin-1 receptor antagonist in ischemic stroke) phase II trial, using subcutaneous (s.c.) injections of IL-1Ra in combination with i.v. thrombolysis, showed reduced plasma IL-6 levels, whereas neurological recovery three months after stroke was unaffected [159]. Exploratory efficacy analysis suggested that the expected beneficial effect of IL-1Ra on clinical outcome by reducing inflammation might have been counteracted by a negative effect, which could represent an interaction with alteplase [159].

Interleukin-6

Another potent proinflammatory cytokine with pleiotropic functions is IL-6, which is expressed on many cell types, including monocytes, neurons and glial cells (Fig. 1j, k)[52, 70]. The pleiotropism of IL-6 may be explained by IL-6 eliciting fundamentally different cellular responses depending on whether the classic or the trans-signaling pathway is activated [152]. This depends on the IL-6 receptor system that consists of the IL-6 receptor (IL-6R) as well as soluble IL-6R (sIL-6R) and glycoprotein 130 (gp130), which due to its cytoplasmic domain is responsible for the signal transduction. Soluble IL-6R is formed by cleavage from the IL-6R by TACE/ADAM17 [141] or by translation of different IL-6R mRNA splice variants [103].
In classic signaling, IL-6 binds to and forms a complex with membrane-bound IL-6R, which then recruits gp130. Trans-signaling occurs when IL-6 binds sIL-6R, which then binds to membrane-anchored gp130 [141]. Unlike IL-6R, which is expressed by neurons, microglia, neutrophils, monocytes, hepatocytes and CD4+ T cells and thus limits classic signaling to only a few tissues [58], gp130 is ubiquitously expressed in the body (reviewed by [145]), increasing the spectrum of IL-6 target cells. Trans-signaling is normally tightly regulated [185] and can be counteracted by a soluble form of gp130 (sgp130), which is generated by alternative splicing of gp130 mRNA and is present in serum [85]. Once IL-6 is released into the blood it can bind sIL-6R but also sgp130 [150], which immediately interferes with IL-6 trans-signaling [58]. As sgp130 levels are much higher than sIL-6R, trans-signaling does not occur under physiological conditions.
Classic IL-6 signaling is believed to be anti-inflammatory and protective [185], while trans-signaling is responsible for the pro-inflammatory effects mediated by IL-6 [147, 152].

Interleukin-6 in experimental stroke

IL-6 is expressed in the normal CNS, where it influences neuronal homeostasis by acting as a neurotrophic factor via the classical signaling pathway (reviewed by [147]). Ischemic stroke in mice and rats leads to a significant increase in the levels of IL-6 from 6 to 12 h (Fig. 1 and 2c), and in both IL-6R and gp130 from 3 days [3, 70]. IL-6 has been shown to be neuroprotective in experimental stroke [192] although this is still debated [29]. In human stroke, IL-6 serum levels increase within the first 24 h and have been shown to correlate significantly with infarct size and survival [11, 157]. A similar correlation has not been observed for sIL-6R [46, 70]. While studies of IL-6 expression in the ischemic brain post-mortem are sparse, one study showed that IL-6 levels were elevated in the infarct already in the acute phase after stroke and remained elevated at later time points [126]. Supporting the neuroprotective effect of brain-derived IL-6 are findings showing a positive effect of IL-6 on post-stroke neurogenesis, leading to long-term functional recovery [111].

Anti-interleukin-6 treatment in ischemic stroke

Similar to patients treated with nonspecific TNF antagonists, non-neurological patients treated with IL-6 inhibitors are at increased risk of infections (reviewed in [169]). Clinical stroke studies show that sIL-6R correlates with the degree of leukocyte infiltration [85] and that sIL-6R neutralizing antibodies are beneficial [146]. In comparison, anti-IL-6R antibodies target both the membrane-bound form of IL-6R and sIL-6R, and therefore, affect classical and trans-signaling equally (Fig. 3c and Table 2).
If classical IL-6 signaling is protective and trans-signaling detrimental, selective neutralization of the potential, detrimental trans-signaling is possible by administration of the chimeric protein sgp130Fc (Fig. 3c and Table 2). Sgp130Fc is a fusion protein that contains the extracellular domain of human gp130 and the Fc-fragment of human IgG1. This allows sgp130Fc to bind to the IL-6/solIL-6R complex, but not to sIL-6R alone [86], whereby spg130Fc blocks trans-signaling [52] (Fig. 2c). Such specific inhibition of the trans-signaling pathway using, i.e. sgp130, which does not compromise classic signaling, could be a promising therapeutic tool in future stroke research.

Interleukin-10 in clinical and experimental stroke

IL-10 is a pleiotropic anti-inflammatory cytokine mainly produced by type-2 helper T cells, which in turn regulate inflammatory reactions. IL-10 binds to IL-10 receptors (IL-10R) to reduce inflammation and limiting apoptosis [148]. In the CNS, astrocytes, neurons, and microglia have been reported to produce IL-10 [114, 188].
A meta-analysis investigating the association of IL10 gene polymorphism with the risk of ischemic stroke showed no overall significant association between IL-10 and the risk of ischemic stroke, but an association was found with large vessel disease and small vessel disease [89], suggesting that some subtypes of ischemic stroke are associated with IL10 gene polymorphisms.
In experimental stroke, IL-10 mRNA and protein and IL-10R mRNA levels are increased, with IL-10 noted in microglia and IL-10R on astrocytes in the peri-infarct area [126, 132]. In transgenic mice overexpressing IL-10, infarct volumes were reduced, and apoptosis decreased 4 days after pMCAO [38]. Furthermore, low IL-10 levels were associated with poor stroke outcome and a delayed, exacerbated inflammatory response after pMCAO that was ameliorated by IL-10 administration after pMCAO [132] (Table 1). Therapeutic administration of IL-10 has been shown to be neuroprotective in experimental stroke and to limit post-stroke inflammation [96, 97, 130, 139, 160, 165] (Table 1),
Low plasma IL-10 levels in patients with subcortical or lacunar stroke are associated with neurological worsening within the first 48 h [180], attributing IL-10 a role in the acute neuroinflammatory response after stroke. This is in line with findings by Protti et al. showing that patients with low IL-10 levels deteriorated neurologically within the first 3 days post-stroke [136]. Stroke patients are prone to infection due to stroke-induced immunodepression, however, and increased serum IL-10 levels have been identified as an independent predictor of post-stroke infection [22, 187]. Women have poorer recovery after ischemic stroke than men, even after controlling for age and stroke severity [19, 80]. This may be partly due to the increased IL-10 levels 24 h post-stroke and an associated higher incidence of post-stroke urinary tract infection and poorer overall outcomes in women have been suggested to be a contributing factor [35]. Overall, these studies indicate that an excessive IL-10 response can lead to post-stroke immunosuppression and worsen neurological outcome, suggesting that IL-10 therapeutics should be given with caution. Future studies should be aimed at differentiating between central and peripheral IL-10 effects post-stroke.

Concluding remarks

The dual role of inflammation in both injury and repair complicates attempts to target inflammatory signals in stroke patients. “Single-target” therapies appear insufficient because ischemic stroke involves several mechanisms. Therapeutic approaches should, therefore, most likely target several cell types and different post-ischemic phases to promote protection and recovery.
A possible new approach is to enhance proinflammatory cytokine inhibition either by simultaneous targeting of more than one cytokine or using a more selective targeting approach where only part of the signaling cascade initiated by a given cytokine is inhibited. More selective targeting can be achieved because some of the detrimental and beneficial signals diverge at the level of ligand (e.g. solTNF or tmTNF and IL-1 or IL-1Ra) and at the level of the receptor (e.g. TNFR1 or TNFR2 and IL-6R or sIL-6R). Accordingly, specific inhibition of solTNF, IL-1, or IL-6 trans-signaling might be sufficient to inhibit the pathological consequences of deregulated cytokine signaling while leaving beneficial signaling pathways intact.
The differential roles of cytokine and cytokine receptors, and the function of cytokines derived from specific cell subsets make it clear that the use of anti-cytokine drugs can be improved or adjusted to the specific disease context. A novel approach to block detrimental inflammation following experimental ischemia is the use of cell-type-restricted targeting of cytokines, or the creation of Activity-on-Target cytokines (AcTakines), which is immunotherapy consisting of mutated cytokines with reduced binding affinity coupled to a targeting moiety that guides cytokines to the desired cell target [60]. Recently, Nedospasov and colleagues designed myeloid cell-specific TNF inhibitors (MYSTIs), which are recombinant mini-antibodies with dual specificity, that can bind to the surface molecule F4/80 or CD11b on myeloid cells and to solTNF and were found to be beneficial in in vivo models of acute hepatotoxicity and arthritis [47, 128].
For anti-inflammatory therapies to be successful in stroke treatment, a better understanding is needed of both the temporal and spatial dynamics of resident microglia and recruited inflammatory cells. Despite intense investigation, there are still numerous controversies concerning the time course of leukocyte recruitment in acute stroke. An improved understanding of the heterogeneity of the inflammatory response in this disease also demands better imaging studies of stroke patients, using tracers to identify both infiltrating cells and functional, relevant cytokine receptors. The heterogenic roles that microglia play in stroke make it challenging to identify strategies that modulate microglial function, but promising results of pre-clinical studies suggest that this should be a major focus of attention in future stroke research.
As evidenced above, post-stroke neuroinflammation is both a tool and a target for therapy. However, care must be taken as to when, where, and how to intervene with neuroinflammatory responses. Taken altogether, this calls for further translational stroke research.

Acknowledgements

Silas Arlt Tvingsholm is acknowledged for help with graphical design and Claire Gudex for proof-reading. This work was supported by the Danish Council for Independent Research, Medical Sciences (DFF-4183-00033 to KLL), the Hoerslev Foundation (BHC), the Lundbeck Foundation (R54-A5539 and R173-2014-955 to KLL, R67-A6383 to BHC and R126-A11512 to BF), and the Novo Nordic Foundation (NNF12OC0002215 to BF).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Aderka D (1996) The potential biological and clinical significance of the soluble tumor necrosis factor receptors. Cytokine Growth Factor Rev 7:231–240CrossRefPubMed Aderka D (1996) The potential biological and clinical significance of the soluble tumor necrosis factor receptors. Cytokine Growth Factor Rev 7:231–240CrossRefPubMed
3.
Zurück zum Zitat Ali C, Nicole O, Docagne F, Lesne S, MacKenzie ET, Nouvelot A et al (2000) Ischemia-induced interleukin-6 as a potential endogenous neuroprotective cytokine against NMDA receptor-mediated excitotoxicity in the brain. J Cereb Blood Flow Metab 20:956–966CrossRefPubMed Ali C, Nicole O, Docagne F, Lesne S, MacKenzie ET, Nouvelot A et al (2000) Ischemia-induced interleukin-6 as a potential endogenous neuroprotective cytokine against NMDA receptor-mediated excitotoxicity in the brain. J Cereb Blood Flow Metab 20:956–966CrossRefPubMed
7.
Zurück zum Zitat Astrup J, Siesjo BK, Symon L (1981) Thresholds in cerebral ischemia - the ischemic penumbra. Stroke 12:723–725CrossRefPubMed Astrup J, Siesjo BK, Symon L (1981) Thresholds in cerebral ischemia - the ischemic penumbra. Stroke 12:723–725CrossRefPubMed
8.
Zurück zum Zitat Barone FC, Arvin B, White RF, Miller A, Webb CL, Willette RN et al (1997) Tumor necrosis factor-alpha. A mediator of focal ischemic brain injury. Stroke 28:1233–1244CrossRefPubMed Barone FC, Arvin B, White RF, Miller A, Webb CL, Willette RN et al (1997) Tumor necrosis factor-alpha. A mediator of focal ischemic brain injury. Stroke 28:1233–1244CrossRefPubMed
14.
Zurück zum Zitat Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson MF et al (1997) A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385:729–733CrossRefPubMed Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson MF et al (1997) A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385:729–733CrossRefPubMed
17.
Zurück zum Zitat Boutin H, LeFeuvre RA, Horai R, Asano M, Iwakura Y, Rothwell NJ (2001) Role of IL-1alpha and IL-1beta in ischemic brain damage. J Neurosci 21:5528–5534CrossRefPubMedPubMedCentral Boutin H, LeFeuvre RA, Horai R, Asano M, Iwakura Y, Rothwell NJ (2001) Role of IL-1alpha and IL-1beta in ischemic brain damage. J Neurosci 21:5528–5534CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Buttini M, Appel K, Sauter A, Gebicke-Haerter PJ, Boddeke HW (1996) Expression of tumor necrosis factor alpha after focal cerebral ischaemia in the rat. Neuroscience 71:1–16CrossRefPubMed Buttini M, Appel K, Sauter A, Gebicke-Haerter PJ, Boddeke HW (1996) Expression of tumor necrosis factor alpha after focal cerebral ischaemia in the rat. Neuroscience 71:1–16CrossRefPubMed
29.
Zurück zum Zitat Clark WM, Rinker LG, Lessov NS, Hazel K, Hill JK, Stenzel-Poore M et al (2000) Lack of interleukin-6 expression is not protective against focal central nervous system ischemia. Stroke 31:1715–1720CrossRefPubMed Clark WM, Rinker LG, Lessov NS, Hazel K, Hill JK, Stenzel-Poore M et al (2000) Lack of interleukin-6 expression is not protective against focal central nervous system ischemia. Stroke 31:1715–1720CrossRefPubMed
37.
Zurück zum Zitat Davies CA, Loddick SA, Toulmond S, Stroemer RP, Hunt J, Rothwell NJ (1999) The progression and topographic distribution of interleukin-1beta expression after permanent middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 19:87–98CrossRefPubMed Davies CA, Loddick SA, Toulmond S, Stroemer RP, Hunt J, Rothwell NJ (1999) The progression and topographic distribution of interleukin-1beta expression after permanent middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 19:87–98CrossRefPubMed
42.
Zurück zum Zitat Dinarello CA (1996) Biologic basis for interleukin-1 in disease. Blood 87:2095–2147PubMed Dinarello CA (1996) Biologic basis for interleukin-1 in disease. Blood 87:2095–2147PubMed
45.
Zurück zum Zitat Dubois CM, Ruscetti FW, Keller JR, Oppenheim JJ, Hestdal K, Chizzonite R et al (1991) In vivo interleukin-1 (IL-1) administration indirectly promotes type II IL-1 receptor expression on hematopoietic bone marrow cells: novel mechanism for the hematopoietic effects of IL-1. Blood 78:2841–2847PubMed Dubois CM, Ruscetti FW, Keller JR, Oppenheim JJ, Hestdal K, Chizzonite R et al (1991) In vivo interleukin-1 (IL-1) administration indirectly promotes type II IL-1 receptor expression on hematopoietic bone marrow cells: novel mechanism for the hematopoietic effects of IL-1. Blood 78:2841–2847PubMed
48.
Zurück zum Zitat Eigenbrod T, Park JH, Harder J, Iwakura Y, Nunez G (2008) Cutting edge: critical role for mesothelial cells in necrosis-induced inflammation through the recognition of IL-1 alpha released from dying cells. J Immunol 181:8194–8198CrossRefPubMed Eigenbrod T, Park JH, Harder J, Iwakura Y, Nunez G (2008) Cutting edge: critical role for mesothelial cells in necrosis-induced inflammation through the recognition of IL-1 alpha released from dying cells. J Immunol 181:8194–8198CrossRefPubMed
54.
Zurück zum Zitat Friedlander RM, Gagliardini V, Hara H, Fink KB, Li W, MacDonald G et al (1997) Expression of a dominant negative mutant of interleukin-1 beta converting enzyme in transgenic mice prevents neuronal cell death induced by trophic factor withdrawal and ischemic brain injury. J Exp Med 185:933–940CrossRefPubMedPubMedCentral Friedlander RM, Gagliardini V, Hara H, Fink KB, Li W, MacDonald G et al (1997) Expression of a dominant negative mutant of interleukin-1 beta converting enzyme in transgenic mice prevents neuronal cell death induced by trophic factor withdrawal and ischemic brain injury. J Exp Med 185:933–940CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Galea J, Ogungbenro K, Hulme S, Greenhalgh A, Aarons L, Scarth S et al (2011) Intravenous anakinra can achieve experimentally effective concentrations in the central nervous system within a therapeutic time window: results of a dose-ranging study. J Cereb Blood Flow Metab 31:439–447. https://doi.org/10.1038/jcbfm.2010.103 CrossRefPubMed Galea J, Ogungbenro K, Hulme S, Greenhalgh A, Aarons L, Scarth S et al (2011) Intravenous anakinra can achieve experimentally effective concentrations in the central nervous system within a therapeutic time window: results of a dose-ranging study. J Cereb Blood Flow Metab 31:439–447. https://​doi.​org/​10.​1038/​jcbfm.​2010.​103 CrossRefPubMed
59.
Zurück zum Zitat Garcia JH, Liu KF, Relton JK (1995) Interleukin-1 receptor antagonist decreases the number of necrotic neurons in rats with middle cerebral artery occlusion. Am J Pathol 147:1477–1486PubMedPubMedCentral Garcia JH, Liu KF, Relton JK (1995) Interleukin-1 receptor antagonist decreases the number of necrotic neurons in rats with middle cerebral artery occlusion. Am J Pathol 147:1477–1486PubMedPubMedCentral
64.
Zurück zum Zitat Granowitz EV, Porat R, Mier JW, Pribble JP, Stiles DM, Bloedow DC et al (1992) Pharmacokinetics, safety and immunomodulatory effects of human recombinant interleukin-1 receptor antagonist in healthy humans. Cytokine 4:353–360CrossRefPubMed Granowitz EV, Porat R, Mier JW, Pribble JP, Stiles DM, Bloedow DC et al (1992) Pharmacokinetics, safety and immunomodulatory effects of human recombinant interleukin-1 receptor antagonist in healthy humans. Cytokine 4:353–360CrossRefPubMed
65.
Zurück zum Zitat Greenfeder SA, Nunes P, Kwee L, Labow M, Chizzonite RA, Ju G (1995) Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex. J Biol Chem 270:13757–13765CrossRefPubMed Greenfeder SA, Nunes P, Kwee L, Labow M, Chizzonite RA, Ju G (1995) Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex. J Biol Chem 270:13757–13765CrossRefPubMed
68.
Zurück zum Zitat Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B et al (1995) The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83:793–802CrossRefPubMed Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B et al (1995) The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83:793–802CrossRefPubMed
69.
Zurück zum Zitat Grell M, Wajant H, Zimmermann G, Scheurich P (1998) The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci USA 95:570–575CrossRefPubMedPubMedCentral Grell M, Wajant H, Zimmermann G, Scheurich P (1998) The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci USA 95:570–575CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z et al (1997) Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci USA 94:2007–2012CrossRefPubMedPubMedCentral Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z et al (1997) Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci USA 94:2007–2012CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Hata R, Maeda K, Hermann D, Mies G, Hossmann KA (2000) Evolution of brain infarction after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab 20:937–946CrossRefPubMed Hata R, Maeda K, Hermann D, Mies G, Hossmann KA (2000) Evolution of brain infarction after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab 20:937–946CrossRefPubMed
76.
Zurück zum Zitat Hayward NJ, Elliott PJ, Sawyer SD, Bronson RT, Bartus RT (1996) Lack of evidence for neutrophil participation during infarct formation following focal cerebral ischemia in the rat. Exp Neurol 139:188–202CrossRefPubMed Hayward NJ, Elliott PJ, Sawyer SD, Bronson RT, Bartus RT (1996) Lack of evidence for neutrophil participation during infarct formation following focal cerebral ischemia in the rat. Exp Neurol 139:188–202CrossRefPubMed
81.
Zurück zum Zitat Ito D, Tanaka K, Suzuki S, Dembo T, Fukuuchi Y (2001) Enhanced expression of Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral ischemia in rat brain. Stroke 32:1208–1215CrossRefPubMed Ito D, Tanaka K, Suzuki S, Dembo T, Fukuuchi Y (2001) Enhanced expression of Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral ischemia in rat brain. Stroke 32:1208–1215CrossRefPubMed
85.
Zurück zum Zitat Jones SA, Rose-John S (2002) The role of soluble receptors in cytokine biology: the agonistic properties of the sIL-6R/IL-6 complex. Biochim Biophys Acta 1592:251–263CrossRefPubMed Jones SA, Rose-John S (2002) The role of soluble receptors in cytokine biology: the agonistic properties of the sIL-6R/IL-6 complex. Biochim Biophys Acta 1592:251–263CrossRefPubMed
86.
Zurück zum Zitat Jostock T, Mullberg J, Ozbek S, Atreya R, Blinn G, Voltz N et al (2001) Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem 268:160–167CrossRefPubMed Jostock T, Mullberg J, Ozbek S, Atreya R, Blinn G, Voltz N et al (2001) Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem 268:160–167CrossRefPubMed
90.
99.
Zurück zum Zitat Loddick SA, Rothwell NJ (1996) Neuroprotective effects of human recombinant interleukin-1 receptor antagonist in focal cerebral ischaemia in the rat. J Cereb Blood Flow Metab 16:932–940CrossRefPubMed Loddick SA, Rothwell NJ (1996) Neuroprotective effects of human recombinant interleukin-1 receptor antagonist in focal cerebral ischaemia in the rat. J Cereb Blood Flow Metab 16:932–940CrossRefPubMed
101.
Zurück zum Zitat Loddick SA, Wong ML, Bongiorno PB, Gold PW, Licinio J, Rothwell NJ (1997) Endogenous interleukin-1 receptor antagonist is neuroprotective. Biochem Biophys Res Commun 234:211–215CrossRefPubMed Loddick SA, Wong ML, Bongiorno PB, Gold PW, Licinio J, Rothwell NJ (1997) Endogenous interleukin-1 receptor antagonist is neuroprotective. Biochem Biophys Res Commun 234:211–215CrossRefPubMed
103.
Zurück zum Zitat Lust JA, Donovan KA, Kline MP, Greipp PR, Kyle RA, Maihle NJ (1992) Isolation of an mRNA encoding a soluble form of the human interleukin-6 receptor. Cytokine 4:96–100CrossRefPubMed Lust JA, Donovan KA, Kline MP, Greipp PR, Kyle RA, Maihle NJ (1992) Isolation of an mRNA encoding a soluble form of the human interleukin-6 receptor. Cytokine 4:96–100CrossRefPubMed
104.
Zurück zum Zitat Madsen PM, Clausen BH, Degn M, Thyssen S, Kristensen LK, Svensson M et al (2016) Genetic ablation of soluble tumor necrosis factor with preservation of membrane tumor necrosis factor is associated with neuroprotection after focal cerebral ischemia. J Cereb Blood Flow Metab 36:1553–1569. https://doi.org/10.1177/0271678X15610339 CrossRefPubMed Madsen PM, Clausen BH, Degn M, Thyssen S, Kristensen LK, Svensson M et al (2016) Genetic ablation of soluble tumor necrosis factor with preservation of membrane tumor necrosis factor is associated with neuroprotection after focal cerebral ischemia. J Cereb Blood Flow Metab 36:1553–1569. https://​doi.​org/​10.​1177/​0271678X15610339​ CrossRefPubMed
105.
Zurück zum Zitat Malyak M, Smith MF Jr, Abel AA, Hance KR, Arend WP (1998) The differential production of three forms of IL-1 receptor antagonist by human neutrophils and monocytes. J Immunol 161:2004–2010PubMed Malyak M, Smith MF Jr, Abel AA, Hance KR, Arend WP (1998) The differential production of three forms of IL-1 receptor antagonist by human neutrophils and monocytes. J Immunol 161:2004–2010PubMed
108.
Zurück zum Zitat McCann FE, Perocheau DP, Ruspi G, Blazek K, Davies ML, Feldmann M et al (2014) Selective tumor necrosis factor receptor I blockade is antiinflammatory and reveals immunoregulatory role of tumor necrosis factor receptor II in collagen-induced arthritis. Arthritis Rheumatol 66:2728–2738. https://doi.org/10.1002/art.38755 CrossRefPubMed McCann FE, Perocheau DP, Ruspi G, Blazek K, Davies ML, Feldmann M et al (2014) Selective tumor necrosis factor receptor I blockade is antiinflammatory and reveals immunoregulatory role of tumor necrosis factor receptor II in collagen-induced arthritis. Arthritis Rheumatol 66:2728–2738. https://​doi.​org/​10.​1002/​art.​38755 CrossRefPubMed
110.
114.
Zurück zum Zitat Mesples B, Plaisant F, Gressens P (2003) Effects of interleukin-10 on neonatal excitotoxic brain lesions in mice. Brain Res Dev Brain Res 141:25–32CrossRefPubMed Mesples B, Plaisant F, Gressens P (2003) Effects of interleukin-10 on neonatal excitotoxic brain lesions in mice. Brain Res Dev Brain Res 141:25–32CrossRefPubMed
121.
Zurück zum Zitat Nawashiro H, Martin D, Hallenbeck JM (1997) Neuroprotective effects of TNF binding protein in focal cerebral ischemia. Brain Res 778:265–271CrossRefPubMed Nawashiro H, Martin D, Hallenbeck JM (1997) Neuroprotective effects of TNF binding protein in focal cerebral ischemia. Brain Res 778:265–271CrossRefPubMed
125.
Zurück zum Zitat Neumann J, Riek-Burchardt M, Herz J, Doeppner TR, Konig R, Hutten H et al (2015) 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 129:259–277. https://doi.org/10.1007/s00401-014-1355-2 CrossRefPubMed Neumann J, Riek-Burchardt M, Herz J, Doeppner TR, Konig R, Hutten H et al (2015) 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 129:259–277. https://​doi.​org/​10.​1007/​s00401-014-1355-2 CrossRefPubMed
134.
Zurück zum Zitat Pfeffer K (2003) Biological functions of tumor necrosis factor cytokines and their receptors. Cytokine Growth Factor Rev 14:185–191CrossRefPubMed Pfeffer K (2003) Biological functions of tumor necrosis factor cytokines and their receptors. Cytokine Growth Factor Rev 14:185–191CrossRefPubMed
135.
Zurück zum Zitat Porteu F, Hieblot C (1994) Tumor necrosis factor induces a selective shedding of its p75 receptor from human neutrophils. J Biol Chem 269:2834–2840PubMed Porteu F, Hieblot C (1994) Tumor necrosis factor induces a selective shedding of its p75 receptor from human neutrophils. J Biol Chem 269:2834–2840PubMed
137.
Zurück zum Zitat Relton JK, Martin D, Thompson RC, Russell DA (1996) Peripheral administration of Interleukin-1 Receptor antagonist inhibits brain damage after focal cerebral ischemia in the rat. Exp Neurol 138:206–213CrossRefPubMed Relton JK, Martin D, Thompson RC, Russell DA (1996) Peripheral administration of Interleukin-1 Receptor antagonist inhibits brain damage after focal cerebral ischemia in the rat. Exp Neurol 138:206–213CrossRefPubMed
138.
Zurück zum Zitat Relton JK, Rothwell NJ (1992) Interleukin-1 receptor antagonist inhibits ischaemic and excitotoxic neuronal damage in the rat. Brain Res Bull 29:243–246CrossRefPubMed Relton JK, Rothwell NJ (1992) Interleukin-1 receptor antagonist inhibits ischaemic and excitotoxic neuronal damage in the rat. Brain Res Bull 29:243–246CrossRefPubMed
150.
Zurück zum Zitat Sawada M, Itoh Y, Suzumura A, Marunouchi T (1993) Expression of cytokine receptors in cultured neuronal and glial cells. Neurosci Lett 160:131–134CrossRefPubMed Sawada M, Itoh Y, Suzumura A, Marunouchi T (1993) Expression of cytokine receptors in cultured neuronal and glial cells. Neurosci Lett 160:131–134CrossRefPubMed
153.
Zurück zum Zitat Selvaraj UM, Stowe AM (2017) Long-term T cell responses in the brain after an ischemic stroke. Discov Med 24:323–333PubMedPubMedCentral Selvaraj UM, Stowe AM (2017) Long-term T cell responses in the brain after an ischemic stroke. Discov Med 24:323–333PubMedPubMedCentral
156.
160.
Zurück zum Zitat Spera PA, Ellison JA, Feuerstein GZ, Barone FC (1998) IL-10 reduces rat brain injury following focal stroke. Neurosci Lett 251:189–192CrossRefPubMed Spera PA, Ellison JA, Feuerstein GZ, Barone FC (1998) IL-10 reduces rat brain injury following focal stroke. Neurosci Lett 251:189–192CrossRefPubMed
164.
Zurück zum Zitat Stellwagen D, Malenka RC (2006) Synaptic scaling mediated by glial TNF-alpha. Nature 440:1054–1059CrossRefPubMed Stellwagen D, Malenka RC (2006) Synaptic scaling mediated by glial TNF-alpha. Nature 440:1054–1059CrossRefPubMed
165.
Zurück zum Zitat Strle K, Zhou JH, Shen WH, Broussard SR, Johnson RW, Freund GG et al (2001) Interleukin-10 in the brain. Crit Rev Immunol 21:427–449CrossRefPubMed Strle K, Zhou JH, Shen WH, Broussard SR, Johnson RW, Freund GG et al (2001) Interleukin-10 in the brain. Crit Rev Immunol 21:427–449CrossRefPubMed
166.
Zurück zum Zitat Stylianou E, O’Neill LA, Rawlinson L, Edbrooke MR, Woo P, Saklatvala J (1992) Interleukin 1 induces NF-kappa B through its type I but not its type II receptor in lymphocytes. J Biol Chem 267:15836–15841PubMed Stylianou E, O’Neill LA, Rawlinson L, Edbrooke MR, Woo P, Saklatvala J (1992) Interleukin 1 induces NF-kappa B through its type I but not its type II receptor in lymphocytes. J Biol Chem 267:15836–15841PubMed
175.
Zurück zum Zitat Touzani O, Boutin H, LeFeuvre R, Parker L, Miller A, Luheshi G et al (2002) Interleukin-1 influences ischemic brain damage in the mouse independently of the interleukin-1 type I receptor. J Neurosci 22:38–43CrossRefPubMedPubMedCentral Touzani O, Boutin H, LeFeuvre R, Parker L, Miller A, Luheshi G et al (2002) Interleukin-1 influences ischemic brain damage in the mouse independently of the interleukin-1 type I receptor. J Neurosci 22:38–43CrossRefPubMedPubMedCentral
178.
Zurück zum Zitat Um JY, An NH, Kim HM (2003) TNF-alpha and TNF-beta gene polymorphisms in cerebral infarction. J Mol Neurosci 21:167–171CrossRefPubMed Um JY, An NH, Kim HM (2003) TNF-alpha and TNF-beta gene polymorphisms in cerebral infarction. J Mol Neurosci 21:167–171CrossRefPubMed
181.
Zurück zum Zitat Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T et al (2003) Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci 23:8692–8700CrossRefPubMedPubMedCentral Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T et al (2003) Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci 23:8692–8700CrossRefPubMedPubMedCentral
182.
Zurück zum Zitat Wallach D, Engelmann H, Nophar Y, Aderka D, Kemper O, Hornik V et al (1991) Soluble and cell surface receptors for tumor necrosis factor. Agents Actions Suppl 35:51–57PubMed Wallach D, Engelmann H, Nophar Y, Aderka D, Kemper O, Hornik V et al (1991) Soluble and cell surface receptors for tumor necrosis factor. Agents Actions Suppl 35:51–57PubMed
183.
Zurück zum Zitat Wang X, Li X, Currie RW, Willette RN, Barone FC, Feuerstein GZ (2000) Application of real-time polymerase chain reaction to quantitate induced expression of interleukin-1beta mRNA in ischemic brain tolerance. J Neurosci Res 59:238–246CrossRefPubMed Wang X, Li X, Currie RW, Willette RN, Barone FC, Feuerstein GZ (2000) Application of real-time polymerase chain reaction to quantitate induced expression of interleukin-1beta mRNA in ischemic brain tolerance. J Neurosci Res 59:238–246CrossRefPubMed
184.
Zurück zum Zitat Wijdenes J, Clement C, Klein B, Morel-Fourrier B, Vita N, Ferrara P et al (1991) Human recombinant dimeric IL-6 binds to its receptor as detected by anti-IL-6 monoclonal antibodies. Mol Immunol 28:1183–1192CrossRefPubMed Wijdenes J, Clement C, Klein B, Morel-Fourrier B, Vita N, Ferrara P et al (1991) Human recombinant dimeric IL-6 binds to its receptor as detected by anti-IL-6 monoclonal antibodies. Mol Immunol 28:1183–1192CrossRefPubMed
191.
Zurück zum Zitat Yamasaki Y, Matsuura N, Shozuhara H, Onodera H, Itoyama Y, Kogure K (1995) Interleukin-1 as a pathogenetic mediator of ischemic brain damage in rats. Stroke 26:676–680; discussion 681 Yamasaki Y, Matsuura N, Shozuhara H, Onodera H, Itoyama Y, Kogure K (1995) Interleukin-1 as a pathogenetic mediator of ischemic brain damage in rats. Stroke 26:676–680; discussion 681
Metadaten
Titel
Post-stroke inflammation—target or tool for therapy?
verfasst von
Kate Lykke Lambertsen
Bente Finsen
Bettina Hjelm Clausen
Publikationsdatum
27.11.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 5/2019
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-018-1930-z

Weitere Artikel der Ausgabe 5/2019

Acta Neuropathologica 5/2019 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.