Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Transl Stroke Res ; 13(3): 364-369, 2022 06.
Article En | MEDLINE | ID: mdl-34455571

Experimental evidence has emerged that local platelet activation contributes to inflammation and infarct formation in acute ischemic stroke (AIS) which awaits confirmation in human studies. We conducted a prospective observational study on 258 consecutive patients undergoing mechanical thrombectomy (MT) due to large-vessel-occlusion stroke of the anterior circulation (08/2018-05/2020). Intraprocedural microcatheter aspiration of 1 ml of local (occlusion condition) and systemic arterial blood samples (self-control) was performed according to a prespecified protocol. The samples were analyzed for differential leukocyte counts, platelet counts, and plasma levels of the platelet-derived neutrophil-activating chemokine C-X-C-motif ligand (CXCL) 4 (PF-4), the neutrophil attractant CXCL7 (NAP-2), and myeloperoxidase (MPO). The clinical-biological relevance of these variables was corroborated by specific associations with molecular-cellular, structural-radiological, hemodynamic, and clinical-functional parameters. Seventy consecutive patients fulfilling all predefined criteria entered analysis. Mean local CXCL4 (+ 39%: 571 vs 410 ng/ml, P = .0095) and CXCL7 (+ 9%: 693 vs 636 ng/ml, P = .013) concentrations were higher compared with self-controls. Local platelet counts were lower (- 10%: 347,582 vs 383,284/µl, P = .0052), whereas neutrophil counts were elevated (+ 10%: 6022 vs 5485/µl, P = 0.0027). Correlation analyses revealed associations between local platelet and neutrophil counts (r = 0.27, P = .034), and between CXCL7 and MPO (r = 0.24, P = .048). Local CXCL4 was associated with the angiographic degree of reperfusion following recanalization (r = - 0.2523, P = .0479). Functional outcome at discharge correlated with local MPO concentrations (r = 0.3832, P = .0014) and platelet counts (r = 0.288, P = .0181). This study provides human evidence of cerebral platelet activation and platelet-neutrophil interactions during AIS and points to the relevance of per-ischemic thrombo-inflammatory mechanisms to impaired reperfusion and worse functional outcome following recanalization.


Ischemic Stroke , Stroke , Chemokines , Humans , Inflammation/complications , Neutrophils , Platelet Activation , Stroke/complications , Treatment Outcome
2.
Cell Death Dis ; 13(1): 20, 2021 12 20.
Article En | MEDLINE | ID: mdl-34930895

In ischemic stroke (IS) impairment of the blood-brain barrier (BBB) has an important role in the secondary deterioration of neurological function. BBB disruption is associated with ischemia-induced inflammation, brain edema formation, and hemorrhagic infarct transformation, but the underlying mechanisms are incompletely understood. Dysfunction of endothelial cells (EC) may play a central role in this process. Although neuronal NLR-family pyrin domain-containing protein 3 (NLRP3) inflammasome upregulation is an established trigger of inflammation in IS, the contribution of its expression in EC is unclear. We here used brain EC, exposed them to oxygen and glucose deprivation (OGD) in vitro, and analyzed their survival depending on inflammasome inhibition with the NLRP3-specific drug MCC950. During OGD, EC death could significantly be reduced when targeting NLRP3, concomitant with diminished endothelial NLRP3 expression. Furthermore, MCC950 led to reduced levels of Caspase 1 (p20) and activated Gasdermin D as markers for pyroptosis. Moreover, inflammasome inhibition reduced the secretion of pro-inflammatory chemokines, cytokines, and matrix metalloproteinase-9 (MMP9) in EC. In a translational approach, IS was induced in C57Bl/6 mice by 60 mins transient middle cerebral artery occlusion and 23 hours of reperfusion. Stroke volume, functional outcome, the BBB integrity, and-in good agreement with the in vitro results-MMP9 secretion as well as EC survival improved significantly in MCC950-treated mice. In conclusion, our results establish the NLRP3 inflammasome as a critical pathogenic effector of stroke-induced BBB disruption by activating inflammatory signaling cascades and pyroptosis in brain EC.


Blood-Brain Barrier/metabolism , Brain Infarction/drug therapy , Brain Infarction/metabolism , Cell Hypoxia/drug effects , Endothelial Cells/metabolism , Furans/administration & dosage , Indenes/administration & dosage , Inflammasomes/antagonists & inhibitors , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Protective Agents/administration & dosage , Pyroptosis/drug effects , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Glucose/metabolism , Inflammasomes/metabolism , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Treatment Outcome
3.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article En | MEDLINE | ID: mdl-33514001

Inflammation is crucial in the pathophysiology of stroke and thus a promising therapeutic target. High-frequency stimulation (HFS) of the mesencephalic locomotor region (MLR) reduces perilesional inflammation after photothrombotic stroke (PTS). However, the underlying mechanism is not completely understood. Since distinct neural and immune cells respond to electrical stimulation by releasing acetylcholine, we hypothesize that HFS might trigger the cholinergic anti-inflammatory pathway via activation of the α7 nicotinic acetylcholine receptor (α7nAchR). To test this hypothesis, rats underwent PTS and implantation of a microelectrode into the MLR. Three hours after intervention, either HFS or sham-stimulation of the MLR was applied for 24 h. IFN-γ, TNF-α, and IL-1α were quantified by cytometric bead array. Choline acetyltransferase (ChAT)+ CD4+-cells and α7nAchR+-cells were quantified visually using immunohistochemistry. Phosphorylation of NFĸB, ERK1/2, Akt, and Stat3 was determined by Western blot analyses. IFN-γ, TNF-α, and IL-1α were decreased in the perilesional area of stimulated rats compared to controls. The number of ChAT+ CD4+-cells increased after MLR-HFS, whereas the amount of α7nAchR+-cells was similar in both groups. Phospho-ERK1/2 was reduced significantly in stimulated rats. The present study suggests that MLR-HFS may trigger anti-inflammatory processes within the perilesional area by modulating the cholinergic system, probably via activation of the α7nAchR.


Inflammation/therapy , Neuroimmunomodulation/genetics , Stroke/therapy , alpha7 Nicotinic Acetylcholine Receptor/genetics , Acetylcholine/metabolism , Animals , Choline O-Acetyltransferase/genetics , Disease Models, Animal , Electric Stimulation , Humans , Inflammation/genetics , Inflammation/pathology , Mesencephalon/pathology , Mesencephalon/radiation effects , Neuroimmunomodulation/radiation effects , Rats , Stroke/genetics , Stroke/pathology , Tumor Necrosis Factor-alpha/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
4.
Curr Neurovasc Res ; 17(5): 636-643, 2020.
Article En | MEDLINE | ID: mdl-33261540

BACKGROUND: Gait impairment after stroke is considered as a loss of cerebral function but is also the result of dysfunctional cerebral signals travelling to the spinal motor centres. A therapeutic option to restore disturbed cerebral network activity is deep brain stimulation (DBS). METHODS: A promising target for neuromodulation might be the pedunculopontine tegmental nucleus (PPTg), which contributes to the initiation and control of gait. To test this hypothesis, we trained eighteen rats to cross a horizontal ladder and a wooden beam before inflicting a photothrombosis in the right sensorimotor cortex and implanting a stimulating electrode in the ipsilateral PPTg. RESULTS: Continuous high-frequency DBS (130 Hz; amplitude 55 ± 5 µA) of rats for 10 days yielded no significant improvement of skilled walking when examined with the ladder rung walking test and beam walking test compared to sham-stimulation. CONCLUSION: In contrast to DBS of the cuneiform nucleus, PPTg-stimulation improves neither control of gait nor balance after stroke.


Deep Brain Stimulation/methods , Gait/physiology , Pedunculopontine Tegmental Nucleus/physiopathology , Sensorimotor Cortex/physiopathology , Thrombotic Stroke/therapy , Walking/physiology , Animals , Disease Models, Animal , Male , Rats , Rats, Wistar , Thrombotic Stroke/physiopathology , Treatment Outcome
5.
Circ Res ; 127(8): 1023-1035, 2020 09 25.
Article En | MEDLINE | ID: mdl-32762491

RATIONALE: Ischemic stroke is a leading cause of morbidity and mortality worldwide. Recanalization of the occluded vessel is essential but not sufficient to guarantee brain salvage. Experimental and clinical data suggest that infarcts often develop further due to a thromboinflammatory process critically involving platelets and T cells, but the underlying mechanisms are unknown. OBJECTIVE: We aimed to determine the role of CD (cluster of differentiation)-84 in acute ischemic stroke after recanalization and to dissect the underlying molecular thromboinflammatory mechanisms. METHODS AND RESULTS: Here, we show that mice lacking CD84-a homophilic immunoreceptor of the SLAM (signaling lymphocyte activation molecule) family-on either platelets or T cells displayed reduced cerebral CD4+ T-cell infiltration and thrombotic activity following experimental stroke resulting in reduced neurological damage. In vitro, platelet-derived soluble CD84 enhanced motility of wild-type but not of Cd84-/- CD4+ T cells suggesting homophilic CD84 interactions to drive this process. Clinically, human arterial blood directly sampled from the ischemic cerebral circulation indicated local shedding of platelet CD84. Moreover, high platelet CD84 expression levels were associated with poor outcome in patients with stroke. CONCLUSIONS: These results establish CD84 as a critical pathogenic effector and thus a potential pharmacological target in ischemic stroke.


Blood Platelets/metabolism , CD4-Positive T-Lymphocytes/metabolism , Infarction, Middle Cerebral Artery/metabolism , Inflammation/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Thrombotic Stroke/metabolism , Aged , Aged, 80 and over , Animals , Blood Coagulation , CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Cytokines/metabolism , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/immunology , Inflammation/genetics , Inflammation/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Prospective Studies , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/genetics , Thrombotic Stroke/genetics , Thrombotic Stroke/immunology
6.
Int J Mol Sci ; 20(16)2019 Aug 19.
Article En | MEDLINE | ID: mdl-31430854

Blood-brain barrier (BBB) disruption is a critical event after ischemic stroke, which results in edema formation and hemorrhagic transformation of infarcted tissue. BBB dysfunction following stroke is partly mediated by proinflammatory agents. We recently have shown that high frequency stimulation of the mesencephalic locomotor region (MLR-HFS) exerts an antiapoptotic and anti-inflammatory effect in the border zone of cerebral photothrombotic stroke in rats. Whether MLR-HFS also has an impact on BBB dysfunction in the early stage of stroke is unknown. In this study, rats were subjected to photothrombotic stroke of the sensorimotor cortex and implantation of a stimulating microelectrode into the ipsilesional MLR. Thereafter, either HFS or sham stimulation of the MLR was applied for 24 h. After scarifying the rats, BBB disruption was assessed by determining albumin extravasation and tight junction integrity (claudin 3, claudin 5, and occludin) using Western blot analyses and immunohistochemistry. In addition, by applying zymography, expression of pro-metalloproteinase-9 (pro-MMP-9) was analyzed. No differences were found regarding infarct size and BBB dysfunction between stimulated and unstimulated animals 24 h after induction of stroke. Our results indicate that MLR-HFS neither improves nor worsens the damaged BBB after stroke. Attenuating cytokines/chemokines in the perilesional area, as mediated by MLR-HFS, tend to play a less significant role in preventing the BBB integrity.


Blood-Brain Barrier/physiopathology , Electric Stimulation Therapy , Mesencephalon/physiopathology , Stroke/therapy , Animals , Male , Rats , Rats, Wistar , Stroke/physiopathology , Tight Junctions/metabolism
...