scholarly journals Adropin confers neuroprotection and promotes functional recovery from ischemic stroke

2021 ◽  
Author(s):  
Changjun Yang ◽  
Bianca P Lavayen ◽  
Lei Liu ◽  
Brian D Sanz ◽  
Kelly M DeMars ◽  
...  

Adropin is a highly-conserved peptide that has been shown to preserve endothelial barrier function. Blood-brain barrier (BBB) disruption is a key pathological event in cerebral ischemia. However, the effects of adropin on ischemic stroke outcomes remain unexplored. Hypothesizing that adropin exerts neuroprotective effects by maintaining BBB integrity, we investigated the role of adropin in stroke pathology utilizing loss- and gain-of-function genetic approaches combined with pharmacological treatment with synthetic adropin peptide. Stroke decreased endogenous adropin levels in the brain and plasma. Adropin treatment or transgenic adropin overexpression robustly reduced brain injury and improved long-term sensorimotor and cognitive function in young and aged mice subjected to ischemic stroke. In contrast, genetic deletion of adropin exacerbated ischemic brain injury. Mechanistically, adropin neuroprotection depends on endothelial nitric oxide synthase and is associated with reduced BBB permeability and neuroinflammation. We identify adropin as a novel neuroprotective peptide with the potential to improve stroke outcomes.

2020 ◽  
Author(s):  
Leon Teo ◽  
Anthony G. Boghdadi ◽  
Jihane Homman-Ludiye ◽  
Iñaki Carril-Mundiñano ◽  
William C. Kwan ◽  
...  

AbstractInfants and adults respond differently to brain injuries. Specifically, improved neuronal sparing along with reduced astrogliosis and glial scarring often observed earlier in life, likely contributes to improved long-term outcomes. Understanding the underlying mechanisms could enable the recapitulation of neuroprotective effects, observed in infants, to benefit adult patients after brain injuries. We reveal that in primates, Eph/ ephrin signaling contributes to age-dependent reactive astrocyte behavior. Ephrin-A5 expression on astrocytes was more protracted in adults, whereas ephrin-A1 was associated only with infant astrocytes. Furthermore, ephrin-A5 exacerbated major hallmarks of astrocyte reactivity via EphA2 and EphA4 receptors, which was subsequently alleviated by ephrin-A1. Rather than suppressing reactivity, ephrin-A1 signaling shifted astrocytes towards GAP43+ neuroprotection, accounting for improved neuronal sparing in infants. Reintroducing ephrin-A1 after middle-aged ischemic stroke significantly attenuated glial scarring, improved neuronal sparing and preserved circuitry. Therefore, beneficial infant mechanisms can be recapitulated in adults to improve outcomes after CNS injuries.


2005 ◽  
Vol 68 (1) ◽  
pp. 226-232 ◽  
Author(s):  
Biljana Musicki ◽  
Hunter C. Champion ◽  
Robyn E. Becker ◽  
Tongyun Liu ◽  
Melissa F. Kramer ◽  
...  

Stroke ◽  
2020 ◽  
Vol 51 (Suppl_1) ◽  
Author(s):  
Yaoming Wang ◽  
Mikko Huuskonen ◽  
Axel Montagne ◽  
Berislav Zlokovic

Pericytes play a key role in maintaining the blood-brain barrier (BBB) integrity. BBB disruption occurs during early stages after ischemic stroke. However, the role of pericytes in the pathogenesis of ischemic stroke remains still understudied. 3K3A-APC, a recombinant variant of activated protein C, has shown benefits in preclinical models of ischemic stroke and has favorable safety profile and reduces hemorrhage in Phase 2 study in ischemic stroke patients (RHAPSODY). In the present study, we used PDGFRβ heterozygous knockout (PDGFRβ+/-) mice to investigate the effects of pericyte deficiency on ischemic brain injury using transient proximal middle cerebral artery occlusion (tMCAO). Additionally, we investigated the effects of 3K3A-APC therapy (0.2mg/kg i.v. 4h after stroke) in this model. Compared to controls, pericyte deficiency in PDGFRβ+/- mice resulted in ~35% increase in the infarct and edema volumes, reduction in pericyte coverage from 58% to 25%, and increased IgG and fibrin deposition suggesting accelerated BBB breakdown 24h after stroke. Additionally, PDGFRβ+/- mice showed by 36% more degenerating Fluoro-Jade+ neurons and exhibited accelerated neurobehavioral abnormalities. 3K3A-APC improved neuropathological changes and functional deficits. Our results suggest that pericyte deficiency worsens brain damage and functional outcome after ischemic stroke in mice suggesting that pericytes may play an important role in protecting brain from post-ischemic. We also suggests that 3K3A-APC protects pericyte function in stroked mice which could contribute to its overall neuroprotective effects.


2016 ◽  
Vol 310 (11) ◽  
pp. H1448-H1454 ◽  
Author(s):  
Ghezal Froogh ◽  
Jun Qin ◽  
Sharath Kandhi ◽  
Yicong Le ◽  
Houli Jiang ◽  
...  

Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid via CYP/epoxygenases, which are catabolized by soluble epoxide hydrolase (sEH) and known to possess cardioprotective properties. To date, the role of sEH in the modulation of pressure-induced myogenic response/constriction in coronary arteries, an important regulatory mechanism in the coronary circulation, and the issue as to whether the disruption of the sEH gene affects the myogenic response sex differentially have never been addressed. To this end, experiments were conducted on male (M) and female (F) wild-type (WT) and sEH-knockout (KO) mice. Pressure-diameter relationships were assessed in isolated and cannulated coronary arteries. All vessels constricted in response to increases in intraluminal pressure from 60 to 120 mmHg. Myogenic vasoconstriction was significantly attenuated, expressed as an upward shift in the pressure-diameter curve of vessels, associated with higher cardiac EETs in M-KO, F-WT, and F-KO mice compared with M-WT controls. Blockade of EETs via exposure of vessels to 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE) prevented the attenuated myogenic constriction in sEH-KO mice. In the presence of 14,15-EEZE, pressure-diameter curves of females presented an upward shift from those of males, exhibiting a sex-different phenotype. Additional administration of Nω-nitro-l-arginine methyl ester eliminated the sex difference in myogenic responses, leading to four overlapped pressure-diameter curves. Cardiac sEH was downregulated in F-WT compared with M-WT mice, whereas expression of endothelial nitric oxide synthase and CYP4A (20-HETE synthase) was comparable among all groups. In summary, in combination with NO, the increased EET bioavailability as a function of genetic deletion and/or downregulation of sEH accounts for the female-favorable attenuation of pressure-induced vasoconstriction.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3620-3620 ◽  
Author(s):  
Frederik Denorme ◽  
Bhanu Kanth Manne ◽  
Yasuhiro Kosaka ◽  
Benjamin Kile ◽  
Matthew T. Rondina ◽  
...  

Background: Anti-platelet agents are used clinically to prevent ischemic stroke but are incompletely effective. Emerging evidence highlights that platelets contribute to ischemic stroke through mechanisms and pathways that are not targeted by classic anti-platelet agents. Besides their role in thrombosis, platelets also mediate inflammation through the formation of heterotypic leukocyte aggregates. In particular, platelet-neutrophil interactions are known to promote brain injury following ischemic stroke. However, the mechanism by which platelets interact with neutrophils and promote thromboinflammation in ischemic stroke remains elusive. Recently, cyclophilin D (CypD)-mediated platelet necrosis emerged as a potential mediator of detrimental platelet-neutrophil interactions. Aims: To investigate the role of CypD-mediated platelet necrosis in the setting of acute ischemic stroke. Methods: Mice harboring a floxed allele of CypD were crossed to animals expressing Cre recombinase controlled by the Pf4 promoter to generate platelet-specific CypD deficient mice (KO). KO mice and littermate controls (WT)were subjected to a transient middle cerebral artery occlusion (tMCAO) model with 1h of cerebral ischemia followed by 23h of reperfusion or a permanent MCAO model with 24h of ischemia. Neurological and motor outcomes and brain infarct size were measured 24h after stroke onset. In addition, we examined both circulating and cerebral platelet-neutrophil aggregates 24h after stroke. Cerebral blood flow was monitored via blood laser doppler flowmetry. Neutrophils were depleted 24h before stroke onset using a neutrophil-depleting antibody to examine the contribution of neutrophils in ischemic stroke. Results: Loss of CypD in platelets significantly improved neurological (p<0.001) and motor (p<0.005) functions and reduced ischemic stroke infarct volume (p<0.0001) after cerebral transient ischemia/reperfusion injury compared to WT controls. To determine whether improved stroke outcomes in KO mice was associated with improved local cerebral blood flow (CBF) during reperfusion, CBF was measured at time points during and after stroke induction. During ischemia, and immediately after reperfusion, CBF was similar in WTand KO mice. Nevertheless, 3h after stroke onset, CBF was significantly greater (48±15% vs 31±10%; p<0.05) in KO mice compared to WTmice. This difference in CBF persisted and was even more pronounced at 24h (54±12% vs 27±8%; p<0.01). To further investigate whether platelet necrosis was contributing to brain infarction during cerebral reperfusion, we employed a permanent MCAO model. With permanent occlusion, no differences were observed in infarct volume, neurological functions, or motor functions between WT or KO mice, suggesting platelet CypD specifically mediates reperfusion injury following transient cerebral ischemia. These detrimental effects of platelet necrosis were attributable, in part, to platelet-neutrophil interactions. Twenty-four hours after stroke, significantly (p<0.01) fewer circulating platelet-neutrophil aggregates were found in KO mice. Underscoring the role of platelet necrosis in this process, we observed that 33±4% of platelets in platelet-neutrophil aggregates expressed phosphatidylserine (PS) on their surface in WTmice. In contrast, only 17.8±5.1% of platelets in platelet-neutrophil aggregates were PS-positive in KO counterparts (p<0.01). Furthermore, KO mice had less neutrophils recruited to their brain relative to WT controls, and cerebral platelet-neutrophil aggregates were virtually absent in KO mice. To determine whether the protective effect observed in KOmice was due to reduced interactions between necrotic platelets and neutrophils, we depleted circulating neutrophils 24h before induction of stroke. Depletion of neutrophils significantly (p<0.05) reduced infarct size and neurological damage following ischemic stroke in WTmice. However, neutrophil depletion conferred no additional protective effect in KOanimals. Conclusions: These results suggest necrotic platelets interact with neutrophils to exacerbate brain injury following ischemic stroke. As inhibiting platelet necrosis does not compromise hemostasis, targeting platelet CypD may be a potential therapeutic strategy to limit brain damage following ischemic stroke. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document