1
|
Leng C, Lin K, Zhou M, Tao X, Sun B, Shu X, Liu W. Apolipoprotein E deficiency exacerbates blood-brain barrier disruption and hyperglycemia-associated hemorrhagic transformation after ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:107987. [PMID: 39218418 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The polymorphism of the apolipoprotein E (ApoE) gene has been implicated in both the susceptibility to neurodegenerative disease and the prognosis of traumatic brain injury (TBI). However, the influence of ApoE on the risk of hemorrhagic transformation (HT) after acute ischemic stroke remains inconclusive. The present study aimed to investigate the potential impact of ApoE deficiency on the risk of hyperglycemia-associated HT and to elucidate the underlying mechanisms. METHODS Wild-type (WT) and ApoE knockout (ApoE-/-) mice were injected with 50 % glucose to induce hyperglycemia and subsequently subjected to 90 min of intraluminal middle cerebral artery occlusion (MCAO). The mortality, neurological function, HT incidence and HT grading-score were evaluated at 24 hours after reperfusion. To evaluate the integrity of blood-brain barrier (BBB), the immunoglobulin G (IgG) leakage and the protein expressions of tight junctions (TJs) were detected using immunofluorescent staining and western blotting. Finally, the levels of matrix metalloproteinases (MMP)-2/9, microglial activation and proinflammatory mediators were investigated using immunofluorescent staining and western blotting. RESULTS ApoE-/- mice exhibited increased mortality and exacerbated neurological impairment, concomitant with more severe hyperglycemia-associated HT 24 hours post-reperfusion. Meanwhile, ApoE deficiency exacerbated the disruption of BBB, characterized by increased leakage of IgG, aggravated degradation of TJs and microvascular basement membranes. Furthermore, ApoE deficiency further aggravated the upregulation of MMP-2/9 and microglia-triggered neuroinflammation. CONCLUSIONS Our findings demonstrate that the absence of ApoE exacerbates neurological impairment and hyperglycemia-associated HT in ischemic stroke mice, which is closely associated with MMP-2/9 signaling and neuroinflammation-mediated disruption of BBB.
Collapse
Affiliation(s)
- Changlong Leng
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Kuan Lin
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Mei Zhou
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Xiaoqin Tao
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China.
| | - Binlian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China.
| | - Xiji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China.
| | - Wei Liu
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China.
| |
Collapse
|
2
|
Safety outcomes of early initiation of antithrombotic agents within 24 h after intravenous alteplase at 0.6 mg/kg. J Neurol Sci 2023; 445:120546. [PMID: 36657370 DOI: 10.1016/j.jns.2023.120546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
BACKGROUND We examined outcome of acute ischemic stroke (AIS) with administration of antithrombotics within 24 h after intravenous low-dose alteplase. METHODS Consecutive AIS patients who were treated with intravenous alteplase at 0.6 mg/kg from 2005 to 2021 were retrospectively included in our single-center registry. Patients were classified into two groups: those who received antithrombotics within 24 h after intravenous alteplase (early initiation group) and those who did not (control group). Safety outcomes were any intracranial hemorrhage (ICH), symptomatic ICH (sICH) within 36 h after onset, and death within 3 months. sICH was defined as any ICH with a ≥ 4-point increase in the National Institutes of Health Stroke Scale (NIHSS) score or death within 36 h. RESULTS Of 1111 patients (women, 426; median age, 76 [interquartile range, 69-83] years; median NIHSS score, 11 [6-19]; cardioembolism, 580 [52.2%]), early initiation group comprised 58 patients (22; 72 [65-80] years; 7 [4-12]; 11 [19.0%]) and control group comprised 1053 patients (404; 77 [69-84] years; 11 [6-19]; 569 [54.1%]). No significant between-group differences were observed in the incidence of any ICH (17.2% vs. 21.6%; adjusted odds ratio [aOR], 1.18; 95% confidence interval [CI], 0.57-2.44), sICH (0% vs. 0.9%, P = 1.00), or death within 3 months (5.2% vs. 6.7%; aOR, 1.23; 95% CI, 0.36-4.23). CONCLUSIONS Early initiation of antithrombotics after intravenous alteplase at 0.6 mg/kg did not increase the rate of sICH or death within 3 months and may be used with caution in patients with advanced neurological deterioration.
Collapse
|
3
|
Abdul Y, Jamil S, Li W, Ergul A. Cerebral microvascular matrix metalloproteinase-3 (MMP3) contributes to vascular injury after stroke in female diabetic rats. Neurochem Int 2023; 162:105462. [PMID: 36509234 PMCID: PMC9839584 DOI: 10.1016/j.neuint.2022.105462] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
Diabetes exacerbates hemorrhagic transformation (HT) after stroke and worsens clinical outcomes. Female patients with diabetes are at a greater risk of stroke and worsened recovery. We have shown that activation of matrix metalloprotease 3 (MMP3) in hyperglycemic settings mediates HT in male rats. In light of our recent findings that diabetic female rats develop greater HT, the current study was designed to test the hypotheses that: 1) cerebral microvascular MMP3 activation contributes to poor functional outcomes and increased hemorrhagic transformations (HT) after ischemic stroke, and 2) MMP3 inhibition can improve functional outcomes in female diabetic rats. Female control and diabetic Wistar rats were subjected to 60 min of middle cerebral artery occlusion (MCAO). One cohort of diabetic animals received a single dose of MMP3 inhibitor (UK356618; 15 mg/kg; iv) or vehicle after reperfusion. Neurobehavioral outcomes, brain infarct size, edema, HT, and MMPs were measured in brain tissue. Diabetic rats had significant neurological deficits on Day 3 after stroke. MMP3 expression and enzyme activity were significantly increased in both micro and macro vessels of diabetic animals. MMP3 inhibition improved functional outcomes and reduced brain edema and HT scores. In conclusion, cerebral endothelial MMP3 activation to vascular injury in female diabetic rats. Our findings identify MMP3 as a potential therapeutic target in diabetic stroke.
Collapse
Affiliation(s)
- Yasir Abdul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, USA; Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, USA
| | - Sarah Jamil
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, USA; Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, USA
| | - Weiguo Li
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, USA; Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, USA
| | - Adviye Ergul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, USA; Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, USA.
| |
Collapse
|
4
|
Zhang W, Wang L, Wang R, Duan Z, Wang H. A blockade of microRNA-155 signal pathway has a beneficial effect on neural injury after intracerebral haemorrhage via reduction in neuroinflammation and oxidative stress. Arch Physiol Biochem 2022; 128:1235-1241. [PMID: 32412861 DOI: 10.1080/13813455.2020.1764047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) have important contributions to multiple pathophysiological processes for cellular response to stress and are considered as promising therapeutic targets with respect to drug development due to their small size, relative ease of delivery, and sequence specificity. Thus, in the current report, we examined the effects of inhibiting miRNA-155 (miR-155) on the levels of pro-inflammatory cytokines (PICs), oxidative stress products as well as vascular endothelial growth factor (VEGF) in the parietal cortex and hippocampus of rats following intracerebral haemorrhage (ICH). Real time PCR was used to examine the levels of miR-155 in the parietal cortex and hippocampus of rats; and ELISA to measure IL-1β, IL-6 and TNF-α, oxidative 8-iso PGF2α and 8-OHdG, and VEGF. Additionally, modified neurological Severity Score (mNSS) was examined to indicate neurological function in animals. In results, with induction of ICH, the levels of miR-155 were amplified in the parietal cortex and hippocampus and this was accompanied with increases of IL-1β, IL-6 and TNF-α; and 8-iso PGF2α and 8-OHdG. Intracerebroventricular infusion of miR-155 inhibitor attenuated the elevation of PICs and amplification of oxidative stress products. Interestingly, miR-155 inhibitor promoted VEGF levels. Furthermore, inhibition of miR-155 led to improvement of neurological deficits in ICH rats. In conclusion, miR-155 signal in the parietal cortex and hippocampus is engaged in the processes of neural injury during ICH and blocking central miR-155 pathway plays a beneficial role in regulating neurological function via reduction in PICs and products of oxidative stress; and enhancement of VEGF. This has implications to target miR-155 and its downstream signal pathway for neuronal dysfunction and vulnerability related to ICH.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Luping Wang
- Department of Anesthesiology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Ruimin Wang
- Department of Operation Room, The Second Hospital of Jilin University, Changchun, China
| | - Zongsheng Duan
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Hushan Wang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Müller S, Kufner A, Dell'Orco A, Rackoll T, Mekle R, Piper SK, Fiebach JB, Villringer K, Flöel A, Endres M, Ebinger M, Nave AH. Evolution of Blood-Brain Barrier Permeability in Subacute Ischemic Stroke and Associations With Serum Biomarkers and Functional Outcome. Front Neurol 2021; 12:730923. [PMID: 34744972 PMCID: PMC8567961 DOI: 10.3389/fneur.2021.730923] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: In the setting of acute ischemic stroke, increased blood-brain barrier permeability (BBBP) as a sign of injury is believed to be associated with increased risk of poor outcome. Pre-clinical studies show that selected serum biomarkers including C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNFα), matrix metallopeptidases (MMP), and vascular endothelial growth factors (VEGFs) may play a role in BBBP post-stroke. In the subacute phase of stroke, increased BBBP may also be caused by regenerative mechanisms such as vascular remodeling and therefore may improve functional recovery. Our aim was to investigate the evolution of BBBP in ischemic stroke using contrast-enhanced (CE) magnetic resonance imaging (MRI) and to analyze potential associations with blood-derived biomarkers as well as functional recovery in subacute ischemic stroke patients. Methods: This is an exploratory analysis of subacute ischemic stroke patients enrolled in the BAPTISe study nested within the randomized controlled PHYS-STROKE trial (interventions: 4 weeks of aerobic fitness training vs. relaxation). Patients with at least one CE-MRI before (v1) or after (v2) the intervention were eligible for this analysis. The prevalence of increased BBBP was visually assessed on T1-weighted MR-images based on extent of contrast-agent enhancement within the ischemic lesion. The intensity of increased BBBP was assessed semi-quantitatively by normalizing the mean voxel intensity within the region of interest (ROI) to the contralateral hemisphere (“normalized CE-ROI”). Selected serum biomarkers (high-sensitive CRP, IL-6, TNF-α, MMP-9, and VEGF) at v1 (before intervention) were analyzed as continuous and dichotomized variables defined by laboratory cut-off levels. Functional outcome was assessed at 6 months after stroke using the modified Rankin Scale (mRS). Results: Ninety-three patients with a median baseline NIHSS of 9 [IQR 6–12] were included into the analysis. The median time to v1 MRI was 30 days [IQR 18–37], and the median lesion volume on v1 MRI was 4 ml [IQR 1.2–23.4]. Seventy patients (80%) had increased BBBP visible on v1 MRI. After the trial intervention, increased BBBP was still detectable in 52 patients (74%) on v2 MRI. The median time to v2 MRI was 56 days [IQR 46–67]. The presence of increased BBBP on v1 MRI was associated with larger lesion volumes and more severe strokes. Aerobic fitness training did not influence the increase of BBBP evaluated at v2. In linear mixed models, the time from stroke onset to MRI was inversely associated with normalized CE-ROI (coefficient −0.002, Standard Error 0.007, p < 0.01). Selected serum biomarkers were not associated with the presence or evolution of increased BBBP. Multivariable regression analysis did not identify the occurrence or evolution of increased BBBP as an independent predictor of favorable functional outcome post-stroke. Conclusion: In patients with moderate-to-severe subacute stroke, three out of four patients demonstrated increased BBB permeability, which decreased over time. The presence of increased BBBP was associated with larger lesion volumes and more severe strokes. We could not detect an association between selected serum biomarkers of inflammation and an increased BBBP in this cohort. No clear association with favorable functional outcome was observed. Trial registration: NCT01954797.
Collapse
Affiliation(s)
- Sarah Müller
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anna Kufner
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Klinik und Hochschulambulanz für Neurologie - Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Andrea Dell'Orco
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Torsten Rackoll
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,BIH QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany.,ExcellenceCluster NeuroCure, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Ralf Mekle
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Sophie K Piper
- Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen B Fiebach
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Kersten Villringer
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Klinik und Hochschulambulanz für Neurologie - Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,ExcellenceCluster NeuroCure, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Martin Ebinger
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Medical Park Berlin Humboldtmühle, Berlin, Germany
| | - Alexander H Nave
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Klinik und Hochschulambulanz für Neurologie - Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
6
|
Jiang Y, Han J, Spencer P, Li Y, Vodovoz SJ, Ning MM, Liu N, Wang X, Dumont AS. Diabetes mellitus: A common comorbidity increasing hemorrhagic transformation after tPA thrombolytic therapy for ischemic stroke. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
7
|
Diabetes Mellitus/Poststroke Hyperglycemia: a Detrimental Factor for tPA Thrombolytic Stroke Therapy. Transl Stroke Res 2020; 12:416-427. [PMID: 33140258 DOI: 10.1007/s12975-020-00872-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Intravenous administration of tissue-type plasminogen activator (IV tPA) therapy has long been considered a mainstay in ischemic stroke management. However, patients respond to IV tPA therapy unequally with some subsets of patients having worsened outcomes after treatment. In particular, diabetes mellitus (DM) is recognized as a clinically important vascular comorbidity that leads to lower recanalization rates and increased risks of hemorrhagic transformation (HT). In this short-review, we summarize the recent advances in understanding of the underlying mechanisms involved in post-IV tPA worsening of outcome in diabetic stroke. Potential pathologic factors that are related to the suboptimal tPA recanalization in diabetic stroke include higher plasma plasminogen activator inhibitor (PAI)-1 level, diabetic atherogenic vascular damage, glycation of the tPA receptor annexin A2, and alterations in fibrin clot density. While factors contributing to the exacerbation of HT in diabetic stroke include hyperglycemia, vascular oxidative stress, and inflammation, tPA neurovascular toxicity and imbalance in extracellular proteolysis are discussed. Besides, impaired collaterals in DM also compromise the efficacy of IV tPA therapy. Additionally, several tPA combination approaches developed from experimental studies that may help to optimize IV tPA therapy are also briefly summarized. In summary, more research efforts are needed to improve the safety and efficacy of IV tPA therapy in ischemic stroke patients with DM/poststroke hyperglycemia.
Collapse
|
8
|
Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood–brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res 2018; 52:1220-1239. [PMID: 30468092 DOI: 10.1080/10715762.2018.1521519] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| | - Xi Chen
- Department of Core Facility, the People’s Hospital of Bao-an Shenzhen, Shenzhen, PR China
- The 8th People’s Hospital of Shenzhen, the Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, PR China
| | - Yunhao Luo
- School of Chinese Medicine, the University of Hong Kong, PR China
| | - Jiangang Shen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
9
|
Lu D, Mai HC, Liang YB, Xu BD, Xu AD, Zhang YS. Beneficial Role of Rosuvastatin in Blood-Brain Barrier Damage Following Experimental Ischemic Stroke. Front Pharmacol 2018; 9:926. [PMID: 30186167 PMCID: PMC6110873 DOI: 10.3389/fphar.2018.00926] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022] Open
Abstract
Hemorrhage transformation is the most challenging preventable complication in thrombolytic therapy and is related to recombinant tissue plasminogen activator (rt-PA)-induced blood–brain barrier (BBB) damage. Intraperitoneal injections of normal or high doses of rosuvastatin were administered to Balb/c mice 20 min prior to middle cerebral artery occlusion (MCAO) surgery for 3 h followed by reperfusion with rt-PA thrombolytic therapy and cerebral blood flow monitoring to investigate whether a high or normal dose of rosuvastatin reduces BBB damage after brain ischemia and rt-PA reperfusion. The integrity of the BBB was ameliorated by normal and high doses of rosuvastatin as determined from Evans blue staining, ultrastructure assessments and immunochemistry at 24 h after reperfusion. The levels of TJ proteins were preserved, potentially by targeting platelet-derived growth factor receptor α (PDGFR-α) and low-density lipoprotein receptor-related protein 1 (LRP1) to inhibit the expression of matrix metalloproteinase proteins (MMPs) by reducing the levels of phosphorylated c-jun-N-terminal kinase (pJNK), phosphorylated mitogen-activated protein kinase (MAPK) p38 (pP38) and increasing the levels of phosphorylated extracellular regulated protein kinases (pERK), and tissue inhibitor of metalloproteinases (TIMPs), as inferred from Western blotting and molecular docking analyses. In summary, rosuvastatin reduced rt-PA therapy-associated BBB permeability by PDGFR-α- and LRP1-associated MAPK pathways to reduce the mortality of mice, and a normal dose of rosuvastatin exerted greater preventative effects on reducing BBB damage than did a high dose in the time window of thrombolytic therapy.
Collapse
Affiliation(s)
- Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| | - Hong-Cheng Mai
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| | - Yu-Bin Liang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| | - Bing-Dong Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| | - An-Ding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| | - Yu-Sheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Fabian RH, Derry PJ, Rea HC, Dalmeida WV, Nilewski LG, Sikkema WKA, Mandava P, Tsai AL, Mendoza K, Berka V, Tour JM, Kent TA. Efficacy of Novel Carbon Nanoparticle Antioxidant Therapy in a Severe Model of Reversible Middle Cerebral Artery Stroke in Acutely Hyperglycemic Rats. Front Neurol 2018; 9:199. [PMID: 29686642 PMCID: PMC5900022 DOI: 10.3389/fneur.2018.00199] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/14/2018] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION While oxidative stress can be measured during transient cerebral ischemia, antioxidant therapies for ischemic stroke have been clinically unsuccessful. Many antioxidants are limited in their range and/or capacity for quenching radicals and can generate toxic intermediates overwhelming depleted endogenous protection. We developed a new antioxidant class, 40 nm × 2 nm carbon nanoparticles, hydrophilic carbon clusters, conjugated to poly(ethylene glycol) termed PEG-HCCs. These particles are high-capacity superoxide dismutase mimics, are effective against hydroxyl radical, and restore the balance between nitric oxide and superoxide in the vasculature. Here, we report the effects of PEG-HCCs administered during reperfusion after transient middle cerebral artery occlusion (tMCAO) by suture in the rat under hyperglycemic conditions. Hyperglycemia occurs in one-third of stroke patients and worsens clinical outcome. In animal models, this worsening occurs largely by accelerating elaboration of reactive oxygen species (ROS) during reperfusion. METHODS PEG-HCCs were studied for their protective ability against hydrogen peroxide in b.End3 brain endothelial cell line and E17 primary cortical neuron cultures. In vivo, hyperglycemia was induced by streptozotocin injection 2 days before tMCAO. 58 Male Sprague-Dawley rats were analyzed. They were injected IV with PBS or PEG-HCCs (4 mg/kg 2×) at the time of recanalization after either 90- or 120-min occlusion. Rats were survived for up to 3 days, and infarct volume characteristics and neurological functional outcome (modified Bederson Score) were assessed. RESULTS PEG-HCCs were protective against hydrogen peroxide in both culture models. In vivo improvement was found after PEG-HCCs with 90-min ischemia with reduction in infarct size (42%), hemisphere swelling (46%), hemorrhage score (53%), and improvement in Bederson score (70%) (p = 0.068-0.001). Early high mortality in the 2-h in the PBS control group precluded detailed analysis, but a trend was found in improvement in all factors, e.g., reduction in infarct volume (48%; p = 0.034) and a 56% improvement in Bederson score (p = 0.055) with PEG-HCCs. CONCLUSION This nano-antioxidant showed some improvement in several outcome measures in a severe model of tMCAO when administered at a clinically relevant time point. Long-term studies and additional models are required to assess potential for clinical use, especially for patients hyperglycemic at the time of their stroke, as these patients have the worst outcomes.
Collapse
Affiliation(s)
- Roderic H. Fabian
- Department of Neurology, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Paul J. Derry
- Department of Neurology and Center for Translational Research on Inflammatory Diseases, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Harriett Charmaine Rea
- Department of Neurology, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - William V. Dalmeida
- Department of Neurology, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | | | | | - Pitchaiah Mandava
- Department of Neurology, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States
| | - Kimberly Mendoza
- Department of Chemistry, Rice University, Houston, TX, United States
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Vladimir Berka
- Division of Hematology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States
| | - James M. Tour
- Departments of Chemistry, Computer Science, Materials Science and NanoEngineering, Smalley-Curl Institute and the NanoCarbon Center, Rice University, Houston, TX, United States
| | - Thomas A. Kent
- Department of Neurology and Center for Translational Research on Inflammatory Diseases, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| |
Collapse
|
11
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 575] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
12
|
Ji J, Xiang P, Li T, Lan L, Xu X, Lu G, Ji H, Zhang Y, Li Y. NOSH-NBP, a Novel Nitric Oxide and Hydrogen Sulfide- Releasing Hybrid, Attenuates Ischemic Stroke-Induced Neuroinflammatory Injury by Modulating Microglia Polarization. Front Cell Neurosci 2017; 11:154. [PMID: 28603491 PMCID: PMC5445131 DOI: 10.3389/fncel.2017.00154] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/11/2017] [Indexed: 12/13/2022] Open
Abstract
NOSH-NBP, a novel nitric oxide (NO) and hydrogen sulfide (H2S)-releasing hybrid, protects brain from ischemic stroke. This study mainly aimed to investigate the therapeutic effect of NOSH-NBP on ischemic stroke and the underlying mechanisms. In vivo, transient middle cerebral artery occlusion (tMCAO) was performed in C57BL/6 mice, with NO-NBP and H2S-NBP as controls. NO and H2S scavengers, carboxy-PTIO and BSS, respectively, were used to quench NO and H2S of NOSH-NBP. In vitro, BV2 microglia/BMDM were induced to the M1/2 phenotype, and conditioned medium (CM) experiments in BV2 microglia, neurons and b.End3 cerebral microvascular endothelial cells (ECs) were performed. Microglial/macrophage activation/polarization was assessed by flow cytometry, Western blot, RT-qPCR, and ELISA. Neuronal and EC survival was measured by TUNEL, flow cytometry, MTT and LDH assays. Transmission electron microscopy, EB extravasation, brain water content, TEER measurement and Western blot were used to detect blood-brain barrier (BBB) integrity and function. Interestingly, NOSH-NBP significantly reduced cerebral infarct volume and ameliorated neurological deficit, with superior effects compared with NO-NBP and/or H2S-NBP in mice after tMCAO. Both NO and H2S-releasing groups contributed to protection by NOSH-NBP. Additionally, NOSH-NBP decreased neuronal death and attenuated BBB dysfunction in tMCAO-treated mice. Furthermore, NOSH-NBP promoted microglia/macrophage switch from an inflammatory M1 phenotype to the protective M2 phenotype in vivo and in vitro. Moreover, the TLR4/MyD88/NF-κB pathway and NLRP3 inflammasome were involved in the inhibitory effects of NOSH-NBP on M1 polarization, while peroxisome proliferator activated receptor gamma signaling contributed to NOSH-NBP induced M2 polarization. These findings indicated that NOSH-NBP is a potential therapeutic agent that preferentially promotes microglial/macrophage M1-M2 switch in ischemic stroke.
Collapse
Affiliation(s)
- Jing Ji
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical UniversityNanjing, China
| | - Pengjun Xiang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical UniversityNanjing, China
| | - Tingting Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical UniversityNanjing, China
| | - Li Lan
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical UniversityNanjing, China
| | - Xiaole Xu
- School of Pharmacy, Nantong UniversityNantong, China
| | - Guo Lu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical UniversityNanjing, China
| | - Hui Ji
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical UniversityNanjing, China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical UniversityNanjing, China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical UniversityNanjing, China
| |
Collapse
|
13
|
Hu Q, Manaenko A, Bian H, Guo Z, Huang JL, Guo ZN, Yang P, Tang J, Zhang JH. Hyperbaric Oxygen Reduces Infarction Volume and Hemorrhagic Transformation Through ATP/NAD +/Sirt1 Pathway in Hyperglycemic Middle Cerebral Artery Occlusion Rats. Stroke 2017; 48:1655-1664. [PMID: 28495827 DOI: 10.1161/strokeaha.116.015753] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/28/2017] [Accepted: 03/31/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Energy depletion is a critical factor leading to cell death and brain dysfunction after ischemic stroke. In this study, we investigated whether energy depletion is involved in hyperglycemia-induced hemorrhagic transformation after ischemic stroke and determined the pathway underlying the beneficial effects of hyperbaric oxygen (HBO). METHODS After 2-hour middle cerebral artery occlusion, hyperglycemia was induced by injecting 50% dextrose (6 mL/kg) intraperitoneally at the onset of reperfusion. Immediately after it, rats were exposed to HBO at 2 atmospheres absolutes for 1 hour. ATP synthase inhibitor oligomycin A, nicotinamide phosphoribosyl transferase inhibitor FK866, or silent mating type information regulation 2 homolog 1 siRNA was administrated for interventions. Infarct volume, hemorrhagic volume, and neurobehavioral deficits were recorded; the level of blood glucose, ATP, and nicotinamide adenine dinucleotide and the activity of nicotinamide phosphoribosyl transferase were monitored; the expression of silent mating type information regulation 2 homolog 1, acetylated p53, acetylated nuclear factor-κB, and cleaved caspase 3 were detected by Western blots; and the activity of matrix metalloproteinase-9 was assayed by zymography. RESULTS Hyperglycemia deteriorated energy metabolism and reduced the level of ATP and nicotinamide adenine dinucleotide and exaggerated hemorrhagic transformation, blood-brain barrier disruption, and neurological deficits after middle cerebral artery occlusion. HBO treatment increased the levels of the ATP and nicotinamide adenine dinucleotide and consequently increased silent mating type information regulation 2 homolog 1, resulting in attenuation of hemorrhagic transformation, brain infarction, as well as improvement of neurological function in hyperglycemic middle cerebral artery occlusion rats. CONCLUSIONS HBO induced activation of ATP/nicotinamide adenine dinucleotide/silent mating type information regulation 2 homolog 1 pathway and protected blood-brain barrier in hyperglycemic middle cerebral artery occlusion rats. HBO might be promising approach for treatment of acute ischemic stroke patients, especially patients with diabetes mellitus or treated with r-tPA (recombinant tissue-type plasminogen activator).
Collapse
Affiliation(s)
- Qin Hu
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Anatol Manaenko
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Hetao Bian
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Zongduo Guo
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Jun-Long Huang
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Zhen-Ni Guo
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Peng Yang
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - Jiping Tang
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.)
| | - John H Zhang
- From the Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, China (Q.H., J.-L.H.); Departments of Physiology and Pharmacology (Q.H., H.B., Z.G., Z.-N.G., P.Y., J.T., J.H.Z.) and Department of Anesthesiology (J.H.Z.), Loma Linda University School of Medicine, CA; and Department of Neurology, University of Erlangen-Nuremberg, Germany (A.M.).
| |
Collapse
|
14
|
Fan Z, Yuan Y, Wang F, Qi Y, Han H, Wu J, Zhang G, Yang L. Diabetes mitigates the recovery following intracranial hemorrhage in rats. Behav Brain Res 2016; 320:412-419. [PMID: 27818237 DOI: 10.1016/j.bbr.2016.10.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/24/2016] [Accepted: 10/29/2016] [Indexed: 10/20/2022]
Abstract
Intracranial hemorrhage (ICH) is a common subtype of stroke with high morbidity and mortality. However, few studies have examined the effects of diabetes on the recovery from ICH-induced brain injury. Therefore, we examined the effects of diabetes on protein levels of aquaporins, neuronal loss, angiogenesis, blood brain barrier (BBB) integrity, and neurological deficits following intra-DH collagenase-induced ICH in the hippocampus. We found that diabetic rats exhibited enhanced AQP9 expression in the hippocampus relative to non-diabetic rats, which was associated with increased behavioral deficits. Additionally, ICH induced neovascularization, proliferation of brain microvascular endothelial cells, and hippocampal neuronal loss. However, ICH-induced neovascularization and proliferation of brain microvascular endothelial cells was severely impaired in diabetic rats. Furthermore, ICH-induced hippocampal neuronal loss was exaggerated in diabetic rats. Finally, ICH impaired BBB integrity in the ipsilateral hemisphere, which was increased in diabetic rats. Taken together, the attenuated brain angiogenesis, increased hippocampal neuronal loss, and impaired BBB integrity in diabetic rats after ICH were associated with enhanced AQP9 expression. This may suggest that AQP9 is one of the underlying mechanisms that can mitigate the recovery from ICH in diabetic populations.
Collapse
Affiliation(s)
- Zhenzeng Fan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Yunchao Yuan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Feng Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Yuepeng Qi
- Department of Neurosurgery, The Hospital of Pingshan County, Shijiazhuang 050000, China
| | - Haie Han
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Jianliang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Gengshen Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Lijun Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| |
Collapse
|
15
|
Kalani A, Chaturvedi P, Kamat PK, Maldonado C, Bauer P, Joshua IG, Tyagi SC, Tyagi N. Curcumin-loaded embryonic stem cell exosomes restored neurovascular unit following ischemia-reperfusion injury. Int J Biochem Cell Biol 2016; 79:360-369. [PMID: 27594413 PMCID: PMC5067233 DOI: 10.1016/j.biocel.2016.09.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/22/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022]
Abstract
We tested whether the combined nano-formulation, prepared with curcumin (anti-inflammatory and neuroprotective molecule) and embryonic stem cell exosomes (MESC-exocur), restored neurovascular loss following an ischemia reperfusion (IR) injury in mice. IR-injury was created in 8-10 weeks old mice and divided into two groups. Out of two IR-injured groups, one group received intranasal administration of MESC-exocur for 7days. Similarly, two sham groups were made and one group received MESC-exocur treatment. The study determined that MESC-exocur treatment reduced neurological score, infarct volume and edema following IR-injury. As compared to untreated IR group, MESC-exocur treated-IR group showed reduced inflammation and N-methyl-d-aspartate receptor expression. Treatment of MESC-exocur also reduced astrocytic GFAP expression and alleviated the expression of NeuN positive neurons in IR-injured mice. In addition, MESC-exocur treatment restored vascular endothelial tight (claudin-5 and occludin) and adherent (VE-cadherin) junction proteins in IR-injured mice as compared to untreated IR-injured mice. These results suggest that combining the potentials of embryonic stem cell exosomes and curcumin can help neurovascular restoration following ischemia-reperfusion injury in mice.
Collapse
Affiliation(s)
- Anuradha Kalani
- Department of Physiology, School of Medicine, University of Louisville, KY, USA.
| | - Pankaj Chaturvedi
- Department of Physiology, School of Medicine, University of Louisville, KY, USA
| | - Pradip K Kamat
- Department of Physiology, School of Medicine, University of Louisville, KY, USA
| | - Claudio Maldonado
- Department of Physiology, School of Medicine, University of Louisville, KY, USA; Department of Surgery, University of Louisville, KY, USA
| | - Philip Bauer
- Department of Surgery, University of Louisville, KY, USA
| | - Irving G Joshua
- Department of Physiology, School of Medicine, University of Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, KY, USA
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, University of Louisville, KY, USA
| |
Collapse
|
16
|
Tan Z, Lucke-Wold BP, Logsdon AF, Turner RC, Tan C, Li X, Hongpaison J, Alkon DL, Simpkins JW, Rosen CL, Huber JD. Bryostatin extends tPA time window to 6 h following middle cerebral artery occlusion in aged female rats. Eur J Pharmacol 2015; 764:404-412. [PMID: 26189021 PMCID: PMC4698807 DOI: 10.1016/j.ejphar.2015.07.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 02/08/2023]
Abstract
Blood-brain barrier (BBB) disruption and hemorrhagic transformation (HT) following ischemic/reperfusion injury contributes to post-stroke morbidity and mortality. Bryostatin, a potent protein kinase C (PKC) modulator, has shown promise in treating neurological injury. In the present study, we tested the hypothesis that administration of bryostatin would reduce BBB disruption and HT following acute ischemic stroke; thus, prolonging the time window for administering recombinant tissue plasminogen activator (r-tPA). Acute cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery (MCAO) in 18-20-month-old female rats using an autologous blood clot with delayed r-tPA reperfusion. Bryostatin (or vehicle) was administered at 2 h post-MCAO and r-tPA was administered at 6 h post-MCAO. Functional assessment, lesion volume, and hemispheric swelling measurements were performed at 24 h post-MCAO. Assessment of BBB permeability, measurement of hemoglobin, assessment of matrix metalloproteinase (MMP) levels by gel zymography, and measurement of PKCε, PKCα, PKCδ expression by western blot were conducted at 24 h post-MCAO. Rats treated with bryostatin prior to r-tPA administration had decreased mortality and hemispheric swelling when compared with rats treated with r-tPA alone. Administration of bryostatin also limited BBB disruption and HT and down-regulated MMP-9 expression while up-regulating PKCε expression at 24 h post-MCAO. Bryostatin administration ameliorates BBB disruption and reduces the risk of HT by down-regulating MMP-9 activation and up-regulating PKCε. In this proof-of-concept study, bryostatin treatment lengthened the time-to-treatment window and enhanced the efficacy and safety of thrombolytic therapy.
Collapse
Affiliation(s)
- Zhenjun Tan
- Department of Neurosurgery, School of Medicine, United States
| | | | - Aric F Logsdon
- Department of Basic Pharmaceutical Science, School of Pharmacy, United States
| | - Ryan C Turner
- Department of Neurosurgery, School of Medicine, United States
| | - Cong Tan
- Department of Physiology and Pharmacology, School of Medicine, United States
| | - Xinlan Li
- Department of Neurosurgery, School of Medicine, United States
| | - Jarin Hongpaison
- Blanchette Rockfeller Neuroscience Institute, West Virginia University, United States
| | - Daniel L Alkon
- Blanchette Rockfeller Neuroscience Institute, West Virginia University, United States
| | - James W Simpkins
- Department of Physiology and Pharmacology, School of Medicine, United States
| | - Charles L Rosen
- Department of Neurosurgery, School of Medicine, United States
| | - Jason D Huber
- Department of Basic Pharmaceutical Science, School of Pharmacy, United States.
| |
Collapse
|