Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Eur J Pharmacol ; 893: 173811, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33345851

ABSTRACT

Anti-inflammatory therapy for early brain injury after subarachnoid hemorrhage is a promising treatment for improving the prognosis. HMGB1 is the initiator of early inflammation after subarachnoid hemorrhage. Oleanolic acid is a natural pentacyclic triterpenoid compound with strong anti-inflammatory activity. It can relieve early brain injury in subarachnoid hemorrhage rats, but its mechanism is not very clear. Here, we study the potential mechanism of Oleanolic acid in the treatment of subarachnoid hemorrhage. First, we demonstrated that oleanolic acid alleviated early brain injury after subarachnoid hemorrhage, including improvement of grading score, neurological score, brain edema and permeability of brain blood barrier. Then we found that oleanolic acid could inhibit the transfer of HMGB1 from nucleus to cytoplasm and reduce the level of serum HMGB1. Furthermore, we found that oleanolic acid decreased the acetylation level of HMGB1 by increasing SIRT1 expression rather than by inhibiting JAK/STAT3 pathway. SIRT1 inhibitor sirtinol eliminated all beneficial effects of oleanolic acid on subarachnoid hemorrhage, which indicated that oleanolic acid inhibited the acetylation of HMGB1 by up regulating SIRT1. In addition, oleanolic acid treatment also reduced the levels of TLR4 and apoptosis related factors and reduced neuronal apoptosis after subarachnoid hemorrhage. In summary, our findings suggest that oleanolic acid may activate SIRT1 by acting as an activator of SIRT1, thereby reducing the acetylation of HMGB1, thus playing an anti-inflammatory role to alleviate early brain injury after subarachnoid hemorrhage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Brain/drug effects , HMGB1 Protein/metabolism , Neuroprotective Agents/pharmacology , Oleanolic Acid/pharmacology , Sirtuin 1/metabolism , Subarachnoid Hemorrhage/drug therapy , Acetylation , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/enzymology , Brain/pathology , Brain Edema/drug therapy , Brain Edema/enzymology , Brain Edema/pathology , Capillary Permeability/drug effects , Disease Models, Animal , HMGB1 Protein/genetics , Male , Protein Processing, Post-Translational , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/genetics , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
2.
Biomed Pharmacother ; 133: 111048, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378955

ABSTRACT

Subarachnoid hemorrhage (SAH) is an acute and severe disease with high disability and mortality. Inflammatory reactions have been proven to occur throughout SAH. Extracellular vesicles derived from mesenchymal stem cells (MSCs-EVs) have shown broad potential for the treatment of brain dysfunction and neuroprotective effects through neurogenesis and angiogenesis after stroke. However, the mechanisms of EVs in neuroinflammation during the acute phase of SAH are not well known. Our present study was designed to investigate the effects of MSCs-EVs on neuroinflammation and the polarization regulation of microglia to the M2 phenotype and related signaling pathways after SAH in rats. The SAH model was induced by an improved method of intravascular perforation, and MSCs-EVs were injected via the tail vein. Post-SAH assessments included neurobehavioral tests as well as brain water content, immunohistochemistry, PCR and Western blot analyses. Our results showed that MSCs-EVs alleviated the expression of inflammatory cytokines in the parietal cortex and hippocampus 24 h and 48 h after SAH and that MSCs-EVs inhibited NF-κB and activated AMPK to reduce inflammation after SAH. Furthermore, MSC-EVs regulated the polarization of microglia toward the M2 phenotype by downregulating interleukin-1ß, cluster of differentiation 16, cluster of differentiation 11b, and inducible nitric oxide synthase and upregulating the expression of cluster of differentiation 206 and arginase-1. Additionally, MSCs-EVs inhibited the neuroinflammatory response and had neuroprotective effects in the brain tissues of rats after SAH. This study may support their use as a potential treatment strategy for early SAH in the future.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/enzymology , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Microglia/enzymology , NF-kappa B/metabolism , Subarachnoid Hemorrhage/surgery , Animals , Brain/pathology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Vesicles/enzymology , Male , Mesenchymal Stem Cells/enzymology , Microglia/pathology , Phenotype , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
3.
J Stroke Cerebrovasc Dis ; 29(12): 104986, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32992175

ABSTRACT

BACKGROUND: Early brain injury (EBI) refers to acute brain injury during the first 72 h after subarachnoid hemorrhage (SAH), which is one of the major causes of poor prognosis after SAH. Here, we investigated the effect and the related mechanism of TSG-6 on EBI after SAH. MATERIALS AND METHODS: The Sprague-Dawley rat model of SAH was developed by the endovascular perforation method. TSG-6 (5µg) was administered by an intraventricular injection within 1.5 h after SAH. The effects of TSG-6 on EBI were assessed by neurological score, brain water content (BWC) and TUNEL staining. Immunofluorescence staining was used to assay NF-κB/p-NF-κB expression in microglia. Protein expression levels of heme oxygenase-1 (HO-1), NADPH oxidase 2 (Nox2), Bcl-2, Bax, and cleaved-caspase-3 were measured to investigate the potential mechanism. The enzyme activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the level of reactive oxygen species (ROS) were analyzed using commercially available kits. RESULTS: The results showed that TSG-6 treatment alleviated the neurobehavioral dysfunction and reduced BWC and the number of TUNEL-positive neurons in EBI after SAH. TSG-6 decreased the ROS level and enhanced the enzyme activity of SOD and GSH-Px after SAH. Furthermore TSG-6 inhibited the NF-κB activation, increased the protein expression levels of HO-1 and Bcl-2 and decreased the expression levels of Nox2, Bax, and cleaved-caspase-3. The administration of TSG-6 siRNA abolished the protective effects of TSG-6 on EBI after SAH. CONCLUSION: We found that TSG-6 attenuated oxidative stress and apoptosis in EBI after SAH partly by inhibiting NF-κB and activating HO-1 pathway in brain tissue.


Subject(s)
Antioxidants/administration & dosage , Brain/drug effects , Cell Adhesion Molecules/administration & dosage , Heme Oxygenase (Decyclizing)/metabolism , NADPH Oxidase 2/metabolism , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Injections, Intraventricular , NF-kappa B/metabolism , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology , Time Factors
4.
Acta Neurochir Suppl ; 127: 43-46, 2020.
Article in English | MEDLINE | ID: mdl-31407061

ABSTRACT

Early brain injury is now considered as an important cause of delayed neurological deterioration after aneurysmal subarachnoid hemorrhage (SAH), and neuronal apoptosis is one of the constituents of early brain injury. Caspase family is popular proteases in apoptotic pathways, but there also exist caspase-independent cell death pathways in many pathologic states. In this study, we investigated the ratio of caspase-related and caspase-unrelated neuronal deaths in a mice endovascular perforation SAH model. At 24 h after SAH, about half of neurons in the perforation-side cortex showed increased cleaved caspase-3 immunoreactivity. On the other hand, about half of cleaved caspase-3-immunonegative neurons showed abnormal morphology, suggesting that they were in the process of some sort of cell death in the absence of caspase-3 activity. These findings suggest that both caspase-dependent and caspase-independent signaling pathways may cause neuronal death after SAH.


Subject(s)
Caspases , Subarachnoid Hemorrhage , Animals , Apoptosis , Caspases/metabolism , Mice , Neurons , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/enzymology
5.
J Stroke Cerebrovasc Dis ; 28(12): 104375, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31590996

ABSTRACT

BACKGROUND: Our previous study showed that propofol, one of the widely used anesthetic agents, can attenuate subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) via inhibiting inflammatory and oxidative reaction. However, it is perplexing whether propofol attenuates inflammatory and oxidative reaction through modulating PI3K/Akt pathway. The present study investigated whether PI3K/Akt pathway is involved in propofol's anti-inflammation, antioxidation, and neuroprotection against SAH-induced EBI. MATERIALS AND METHODS: Adult Sprague-Dawley rats underwent SAH and received treatment with propofol or vehicle after 2 and 12 hours of SAH. LY294002 was injected intracerebroventricularly to selectively inhibit PI3K/Akt signaling. Mortality, SAH grading, neurological scores, brain water content, evans blue extravasation, myeloperoxidase, malondialdehyde, superoxide dismutase, and glutathione peroxidase were measured 24 hours after SAH. Immunoreactivity of p-Akt, t-Akt, nuclear factor- kappa B (NF-κB) p65, nuclear factor erythroid-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase (NQO1), and cyclooxygenase-2 (COX-2) in rat brain was determined by western blot. Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in rat brain were examined by ELISA. RESULTS: Propofol significantly reduces neurological dysfunction, BBB permeability, brain edema, inflammation, and oxidative stress, all of which were reversed by LY294002. Propofol significantly upregulates the immunoreactivity of p-Akt, Nrf2, and NQO1, all of which were abolished by LY294002. Propofol significantly downregulates the overexpression of NF-κB p65, COX-2, TNF-α, and IL-1ß, all of which were inhibited by LY294002. CONCLUSION: These results suggest that propofol attenuates SAH-induced EBI by inhibiting inflammatory reaction and oxidative stress, which might be associated with the activation of PI3K/Akt signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Edema/prevention & control , Brain/drug effects , Encephalitis/prevention & control , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Propofol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Subarachnoid Hemorrhage/drug therapy , Animals , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/pathology , Interleukin-1beta/metabolism , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
World Neurosurg ; 132: e795-e801, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31404697

ABSTRACT

OBJECTIVE: To examine changes of expression and activity of phosphodiesterase V (PDE V) in the basilar artery following cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH) in a rabbit model. METHODS: A rabbit model of CVS after SAH was constructed by double blood injection into the cisterna magna. Subjects were divided into 3 groups: blank control group, normal saline group, and SAH group. Transcranial Doppler and selective vertebrobasilar digital subtraction angiography were performed to identify changes of CVS. Changes of PDE V expression and activity were examined. RESULTS: Mean basilar arterial blood flow rate measured by transcranial Doppler was significantly increased in the SAH group compared with the blank control group and normal saline group. Mean basilar artery diameter measured by digital subtraction angiography in the SAH group was narrower than in the other 2 groups. Compared with the other 2 groups, the expression of PDE V in the SAH group was significantly upregulated, and the activity was significantly enhanced. CONCLUSIONS: The rabbit model of SAH-induced CVS was successfully constructed through double blood injection method. Increased basilar artery blood flow, narrowing of the basilar artery, increased PDE V expression, and enhanced PDE V activity in the basilar artery were detected in the CVS rabbits, suggesting that PDE V has the potential to be used as a target for CVS therapy.


Subject(s)
Basilar Artery/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Vasospasm, Intracranial/enzymology , Angiography, Digital Subtraction , Animals , Basilar Artery/diagnostic imaging , Cerebrovascular Circulation , Cisterna Magna , Disease Models, Animal , Immunohistochemistry , Male , Rabbits , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/enzymology , Ultrasonography, Doppler, Transcranial , Vasospasm, Intracranial/diagnostic imaging , Vasospasm, Intracranial/etiology
7.
Stroke ; 50(2): 520-523, 2019 02.
Article in English | MEDLINE | ID: mdl-30602353

ABSTRACT

Background and Purpose- Subarachnoid hemorrhage (SAH) is a devastating form of stroke. Oxidative stress contributes to brain injury, but the mechanisms have been poorly studied. Here, we evaluated the role of 12/15-lipoxygenase (12/15-LOX), an enzyme known to cause cell death in ischemic stroke, on brain injury in a mouse model of SAH. Methods- C57Bl6 wild-type mice and Alox15 knockout mice were subjected to SAH using a direct blood injection technique. In SAH wild-type mice, half received the 12/15-LOX inhibitor ML351 and half received vehicle. Immunohistochemistry, brain edema, blood-brain barrier leakage and functional outcomes were assessed 1 and 3 days after SAH induction. Results- SAH led to increased 12/15-LOX in macrophages of the brain parenchyma, adjacent to the subarachnoid blood. Neuronal cell death after SAH was reduced by ML351 and in Alox15 knockout mice. Similarly, SAH induced brain edema, which was 12/15-LOX dependent. Finally, Alox15 gene knockout and inhibitor treatment in wild-type mice with SAH led to an improved behavioral outcome. Conclusions- 12/15-LOX is overexpressed in macrophages after SAH in mice, and inhibition of the 12/15-LOX pathway decreases brain injury and improves neurological outcome. This study suggests 12/15-LOX as a novel therapeutic target to limit brain injury after SAH.


Subject(s)
Arachidonate 12-Lipoxygenase , Arachidonate 15-Lipoxygenase , Brain Injuries , Isoxazoles/pharmacology , Lipoxygenase Inhibitors/pharmacology , Macrophages , Naphthalenes/pharmacology , Oxidative Stress , Subarachnoid Hemorrhage , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Brain Injuries/drug therapy , Brain Injuries/enzymology , Brain Injuries/genetics , Brain Injuries/pathology , Disease Models, Animal , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/genetics , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology
8.
J Thromb Haemost ; 16(7): 1413-1422, 2018 07.
Article in English | MEDLINE | ID: mdl-29729651

ABSTRACT

Essentials von Willebrand Factor (VWF) and ADAMTS13 may affect early injury after subarachnoid hemorrhage (SAH). Early brain injury was assessed in VWF-/- , ADAMTS13-/- and recombinant (r) ADAMTS13 treated mice. VWF-/- and rADAMTS13 treated mice had less brain injury than ADAMTS13-/- and wild-type mice. Early administration of rADAMTS13 may improve outcome after SAH by reducing early brain injury. SUMMARY: Background Early brain injury is an important determinant of poor functional outcome and case fatality after aneurysmal subarachnoid hemorrhage (SAH), and is associated with early platelet aggregation. No treatment exists for early brain injury after SAH. We investigated whether von Willebrand factor (VWF) is involved in the pathogenesis of early brain injury, and whether ultra-early treatment with recombinant ADAMTS-13 (rADAMTS-13) reduces early brain injury after experimental SAH. Methods Experimental SAH in mice was induced by prechiasmatic injection of non-anticoagulated blood from a littermate. The following experimental SAH groups were investigated: C57BL/6J control (n = 21), VWF-/- (n = 25), ADAMTS-13-/- (n = 23), and C57BL/6J treated with rADAMTS-13 (n = 26). Mice were killed at 2 h after SAH. Primary outcome measures were microglial activation (IBA-1 surface area) and neuronal injury (number of cleaved caspase-3-positive neurons). Results As compared with controls, microglial activation was decreased in VWF-/- mice (mean difference of - 20.0%, 95% confidence interval [CI] - 4.0% to - 38.6%), increased in ADAMTS-13-/- mice (mean difference of + 34.0%, 95% CI 16.2-51.7%), and decreased in rADAMTS-13-treated mice (mean difference of - 22.1%, 95% CI - 3.4% to - 39.1%). As compared with controls (185 neurons, interquartile range [IQR] 133-353), neuronal injury in the cerebral cortex was decreased in VWF-/- mice (63 neurons, IQR 25-78), not changed in ADAMTS-13-/- mice (53 neurons, IQR 26-221), and reduced in rADAMTS-13-treated mice (45 neurons, IQR 9-115). Conclusions Our findings suggest that VWF is involved in the pathogenesis of early brain injury, and support the further study of rADAMTS-13 as a treatment option for early brain injury after SAH.


Subject(s)
ADAMTS13 Protein/metabolism , Brain Injuries/etiology , Brain/enzymology , Subarachnoid Hemorrhage/complications , von Willebrand Factor/metabolism , ADAMTS13 Protein/administration & dosage , ADAMTS13 Protein/deficiency , ADAMTS13 Protein/genetics , Animals , Apoptosis , Brain/drug effects , Brain/pathology , Brain Injuries/enzymology , Brain Injuries/genetics , Brain Injuries/prevention & control , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Disease Models, Animal , Drug Administration Schedule , Female , Genetic Predisposition to Disease , Male , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/administration & dosage , Phenotype , Recombinant Proteins/administration & dosage , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/genetics , Time Factors , von Willebrand Factor/genetics
9.
Cerebrovasc Dis ; 45(3-4): 180-186, 2018.
Article in English | MEDLINE | ID: mdl-29614486

ABSTRACT

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) from intracranial aneurysm rupture results in significant morbidity and mortality. In the present study, we examined the effect of most widely used antiplatelet drugs, aspirin and cilostazol, on aneurysm rupture prevention using a mouse intracranial aneurysm model. MATERIALS AND METHODS: Intracranial aneurysms were induced by a combination of deoxycorticosterone acetate-salt and a single injection of elastase into the cerebrospinal fluid in mice. Treatment with aspirin or cilostazol was started 1 day after aneurysm induction. Aneurysm rupture was detected by neurological symptoms and the presence of intracranial aneurysm with SAH was confirmed by post-mortem examination. RESULTS: Aspirin (10 mg/kg) significantly reduced aneurysm rupture (control:aspirin = 80%:31%, p < 0.05) without affecting the overall incidence of aneurysm formation (60%:62%). Cilostazol (3 mg/kg, 30 mg/kg) did not reduce both rupture rate (control:3 mg/kg:30 mg/kg = 81%:67%:77%) and the overall incidence of aneurysm formation (control:3 mg/kg:30 mg/kg = 72%:71%:76%). Tail vein bleeding time prolonged significantly in both aspirin and cilostazol groups (p < 0.01). CONCLUSION: Aspirin prevented aneurysm rupture in a mouse intracranial aneurysm model, while cilostazol did not. Aspirin, the most frequently used drug for patients with ischemic myocardial and cerebral diseases, is also effective in preventing cerebral aneurysmal rupture.


Subject(s)
Aneurysm, Ruptured/prevention & control , Aspirin/pharmacology , Cerebral Arteries/drug effects , Cilostazol/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Intracranial Aneurysm/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Subarachnoid Hemorrhage/prevention & control , Aneurysm, Ruptured/chemically induced , Aneurysm, Ruptured/enzymology , Aneurysm, Ruptured/pathology , Animals , Cerebral Arteries/enzymology , Cerebral Arteries/pathology , Cyclooxygenase 2/metabolism , Desoxycorticosterone Acetate , Disease Models, Animal , Intracranial Aneurysm/chemically induced , Intracranial Aneurysm/enzymology , Intracranial Aneurysm/pathology , Male , Mice, Inbred C57BL , Pancreatic Elastase , Subarachnoid Hemorrhage/chemically induced , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
10.
J Neurosurg ; 128(1): 86-93, 2018 01.
Article in English | MEDLINE | ID: mdl-28106497

ABSTRACT

OBJECTIVE Renin-angiotensin system (RAS) genetic polymorphisms are thought to play a role in cerebral aneurysm formation and rupture. The Cerebral Aneurysm Renin-Angiotensin System (CARAS) study prospectively evaluated common RAS polymorphisms and their relation to aneurysmal subarachnoid hemorrhage (aSAH). METHODS The CARAS study prospectively enrolled aSAH patients and controls at 2 academic centers in the United States. A blood sample was obtained from all patients for genetic evaluation and measurement of plasma angiotensin-converting enzyme (ACE) concentration. Common RAS polymorphisms were detected using 5' exonuclease (TaqMan) genotyping assays and restriction fragment length polymorphism analysis. RESULTS Two hundred forty-eight patients were screened, and 149 aSAH patients and 50 controls were available for analysis. There was a recessive effect of the C allele of the angiotensinogen ( AGT) C/T single-nucleotide polymorphism (SNP) (OR 1.94, 95% CI 0.912-4.12, p = 0.0853) and a dominant effect of the G allele of the angiotensin II receptor Type 2 ( AT2) G/A SNP (OR 2.11, 95% CI 0.972-4.57, p = 0.0590) on aSAH that did not reach statistical significance after adjustment for potential confounders. The ACE level was significantly lower in aSAH patients with the II genotype (17.6 ± 8.0 U/L) as compared with the ID (22.5 ± 12.1 U/L) and DD genotypes (26.6 ± 14.2 U/L) (p = 0.0195). CONCLUSIONS The AGT C/T and AT2 G/A polymorphisms were not significantly associated with aSAH after controlling for potential confounders. However, a strong trend was identified for a dominant effect of the G allele of the AT2 G/A SNP. Downregulation of the local RAS may contribute to the formation of cerebral aneurysms and subsequent presentation with aSAH. Further studies are required to elucidate the relevant pathophysiology and its potential implication in treatment of patients with aSAH.


Subject(s)
Angiotensinogen/genetics , Intracranial Aneurysm/genetics , Polymorphism, Single Nucleotide , Receptor, Angiotensin, Type 2/genetics , Renin-Angiotensin System/genetics , Subarachnoid Hemorrhage/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Intracranial Aneurysm/enzymology , Intracranial Aneurysm/etiology , Male , Middle Aged , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Prospective Studies , Receptor, Angiotensin, Type 1/genetics , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/enzymology
11.
Nitric Oxide ; 81: 67-74, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29074292

ABSTRACT

INTRODUCTION: We previously demonstrated that cyclic AMP-dependent protein kinase (PKA) phosphorylates neuronal nitric oxide synthase (nNOS) at Ser1412 in the hippocampal dentate gyrus after forebrain ischemia; this phosphorylation event activates NOS activity and might contribute to depression after cerebral ischemia. In this study, we revealed chronological and topographical changes in the phosphorylation of nNOS at Ser1412 immediately after subarachnoid hemorrhage (SAH). METHODS: In a rat single-hemorrhage model of SAH, the hippocampus and adjacent cortex were collected up to 24 h after SAH. Samples from rats that were not injected with autologous blood were used as controls. NOS was partially purified from crude samples via an ADP-agarose gel. Levels of nNOS, nNOS phosphorylated at Ser1412 (p-nNOS), PKA, and p-PKA at Thr197 were studied in the rat hippocampus and cortex using Western blot analyses and immunohistochemistry. RESULTS: According to the Western blot analysis, levels of p-nNOS at Ser1412 were significantly increased in the hippocampus, but not in the cortex, between 1 and 3 h after SAH. Immunohistochemistry revealed the phosphorylation of nNOS at Ser1412 and PKA at Thr197 in the dentate gyrus, but not in the CA1 area, 1 h after SAH. An injection of saline instead of blood also significantly increased levels of p-nNOS at Ser1412 in the hippocampus 1 h after the injection. CONCLUSIONS: An immediate increase in intracranial pressure (ICP) might induce transient cerebral ischemia and promote the PKA-mediated phosphorylation of nNOS at Ser1412 in the dentate gyrus. This signal transduction pathway induces the excessive production of nitric oxide (NO) and might be involved in cognitive dysfunction after SAH.


Subject(s)
Dentate Gyrus/enzymology , Nitric Oxide Synthase Type I/metabolism , Subarachnoid Hemorrhage/enzymology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Dentate Gyrus/metabolism , Male , Neurons/enzymology , Neurons/pathology , Phosphorylation , Rats, Sprague-Dawley , Serine/metabolism , Subarachnoid Hemorrhage/metabolism , Threonine/metabolism
12.
J Neuroinflammation ; 14(1): 137, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28720149

ABSTRACT

BACKGROUND: Heparanase, a mammalian endo-ß-D-glucoronidase that specifically degrades heparan sulfate, has been implicated in inflammation and ischemic stroke. However, the role of heparanase in neuroinflammatory response in subarachnoid hemorrhage (SAH) has not yet been investigated. This study was designed to examine the association between heparanase expression and neuroinflammation during subarachnoid hemorrhage. METHODS: Rats were subjected to SAH by endovascular perforation, and the expression of heparanase was determined by Western blot analysis and immunofluorescence in the ipsilateral brain cortex at 24 h post-SAH. Pial venule leukocyte trafficking was monitored by using intravital microscopy through cranial window. RESULTS: Our results indicated that, compared to their sham-surgical controls, the rats subjected to SAH showed marked elevation of heparanase expression in the ipsilateral brain cortex. The SAH-induced elevation of heparanase was accompanied by increased leukocyte trafficking in pial venules and significant neurological deficiency. Intracerebroventricular application of a selective heparanase inhibitor, OGT2115, which was initiated at 3 h after SAH, significantly suppressed the leukocyte trafficking and improved the neurological function. CONCLUSIONS: Our findings indicate that heparanase plays an important role in mediating the neuroinflammatory response after SAH and contributes to SAH-related neurological deficits and early brain injury following SAH.


Subject(s)
Glucuronidase/biosynthesis , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology , Animals , Inflammation/enzymology , Inflammation/pathology , Male , Random Allocation , Rats , Rats, Sprague-Dawley
13.
Thromb Haemost ; 117(4): 691-699, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28102428

ABSTRACT

Increased von Willebrand factor (VWF) and reduced ADAMTS13 activity are associated with arterial thrombosis. This may also be the culprit mechanism implicated in delayed cerebral ischaemia after aneurysmal subarachnoid haemorrhage (SAH). It was our objective to determine plasma VWF and ADAMTS13 in patients with SAH and healthy subjects; and to explore the levels of those markers and outcome after SAH. Forty consecutive patients were enrolled between September 2007 and April 2014 in a pilot study. Plasma samples were collected from SAH patients on post-bleed day (PBD) 0, 1, 3, 5, 7 and 10 and healthy controls. VWF antigen (VWFAg) and VWF activity (VWFAc) were determined by enzyme-linked immunoassay and collagen binding assay, respectively. ADAMTS13 activity was determined by the cleavage of a fluorescent substrate. Univariate descriptive statistics and cluster analyses were performed based on outcomes in the group with SAH only. Mean age of SAH patients was 52.4 years (26-84 years) and 30 (75 %) were women. 12/40 (30 %) had a high Hunt and Hess grade (IV-V) and 25 (62.5 %) were treated with coil embolisation. Plasma VWFAg and VWFAc were significantly higher in SAH patients than those in healthy subjects on each PBD (p<0.0001). Concurrently, plasma ADAMTS13 activity in SAH patients was significantly lower than that in healthy subjects (p<0.0001). Among those with SAH, cluster analysis demonstrated that patients with higher VWFAg and VWFAc and/or lower ADAMTS13 activity might be at risk of increased mortality. In conclusion, the relative deficiency of plasma ADAMTS13 activity in SAH patients may associate with worse outcome.


Subject(s)
ADAMTS13 Protein/blood , Subarachnoid Hemorrhage/blood , von Willebrand Factor/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Blood Coagulation , Case-Control Studies , Cluster Analysis , Embolization, Therapeutic/instrumentation , Female , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/therapy , Time Factors , Treatment Outcome
14.
Clin Chim Acta ; 460: 196-202, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27380999

ABSTRACT

BACKGROUND: Caspase-3 represents a promising marker of apoptosis. This study was designed to investigate the serial change of serum caspase-3 activities and analyze the relationships between caspase-3 activities and the severity and prognosis of aneurysmal subarachnoid hemorrhage (aSAH). METHODS: We determined serum caspase-3 activities of 118 controls at study entry and 118 patients at admission and at days 1, 2, 3, 5 and 7 after aSAH. Activities were compared with regard to (i) time interval between onset of symptoms and blood sampling, (ii) stroke severity quantified by World Federation of Neurological Surgeons (WFNS) scores and modified Fisher scores and (iii) 6-month outcome. RESULTS: Serum caspase-3 activities were increased after aSAH, peaked at day 3, gradually decreased afterwards, and substantially were higher in patients than in controls. Caspase-3 activities were higher in patients suffering from death or an unfavorable outcome (Glasgow Outcome Scale score of 1-3), had close relation to WFNS scores and modified Fisher scores, and possessed high areas under receiver operating characteristic curve. Moreover, caspase-3 activities at admission and at day 3 predicted poor outcome independently of age, WFNS scores and modified Fisher scores. CONCLUSION: Increased serum caspase-3 activities are highly associated with the severity and prognosis after aSAH.


Subject(s)
Caspase 3/blood , Intracranial Aneurysm/diagnosis , Subarachnoid Hemorrhage/diagnosis , Adult , Age Factors , Aged , Biomarkers/blood , Case-Control Studies , Humans , Intracranial Aneurysm/blood , Intracranial Aneurysm/enzymology , Middle Aged , Prognosis , Severity of Illness Index , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/enzymology , Time Factors
15.
Neuropharmacology ; 108: 316-23, 2016 09.
Article in English | MEDLINE | ID: mdl-27084696

ABSTRACT

BACKGROUND AND PURPOSE: Since tozasertib is neuroprotective for injured optic nerve, this study is intended to test whether tozasertib reduces early brain injury after subarachnoid hemorrhage (SAH) in a rat model. METHODS: Two hundred sixteen (216) male Sprague-Dawley rats were randomly subjected to endovascular perforation model of SAH and sham group. SAH grade, neurological score, and brain water content were measured at 24 and 72 h after SAH. Dual leucine zipper kinase (DLK) and its downstream factors, JNK-interacting protein 3 (JIP3), MA2K7, p-JNK/JNK (c-Jun N-terminal kinase), and apoptosis related proteins cleaved caspase-3 (CC-3), Bim, Bcl-2, and cleaved caspase-9 (CC-9) were analyzed by western blot at 24 h after SAH. Apoptotic cells were detected by terminal deoxynucleotid transferase-deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL). DLK small interfering RNA (siRNA), JIP3 siRNA and MA2K7 siRNA, the JNK, p38MAPK, and MEK inhibitors SP600125, SB203580, and PD98059 were used for intervention. RESULTS: Tozasertib reduced neuronal apoptosis, attenuated brain edema and improved neurobehavioral deficits 24 and 72 h after SAH. At 24 h After SAH, DLK/JIP3/MA2K7/p-JNK/CC-3 expressions were elevated markedly and tozasertib reduced DLK, MA2K7/p-JNK/CC-3 expressions but enhanced JIP3 expression. In the presence of tozasertib, DLK/JIP3/MA2K7 siRNA and SP600125, SB203580 and PD98059 deteriorated the neurobehavioral deficits, brain edema and increased the expression of CC-3. SAH potentiated the expression of Bim, CC-9, and CC-3 but reduced Bcl-2, while tozasertib reduced expression of Bim, CC-9, and CC-3 but enhanced Bcl-2. CONCLUSIONS: Tozasertib reduced neuronal apoptosis and improved outcome possibly via DLK/JIP3/MA2K7/JNK pathways after SAH.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Apoptosis/physiology , Brain Injuries/enzymology , MAP Kinase Kinase Kinases/biosynthesis , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/biosynthesis , Piperazines/administration & dosage , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Injuries/etiology , Brain Injuries/prevention & control , Dose-Response Relationship, Drug , Injections, Intraventricular , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Nerve Tissue Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/prevention & control
16.
Neurosurgery ; 78(4): 487-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26606669

ABSTRACT

BACKGROUND: The value of neuron-specific enolase (NSE) in predicting clinical outcomes has been investigated in a variety of neurological disorders. OBJECTIVE: To investigate the associations of serum NSE with severity of bleeding and functional outcomes in patients with subarachnoid hemorrhage (SAH). METHODS: We retrospectively reviewed the records of patients with SAH from June 2008 to June 2012. The severity of SAH bleeding at admission was measured radiographically with the Fisher scale and clinically with the Glasgow Coma Scale, Hunt and Hess grade, and World Federation of Neurologic Surgeons scale. Outcomes were assessed with the modified Rankin Scale at discharge. RESULTS: We identified 309 patients with nontraumatic SAH, and 71 had NSE testing. Median age was 54 years (range, 23-87 years), and 44% were male. In multivariable analysis, increased NSE was associated with a poorer Hunt and Hess grade (P = .003), World Federation of Neurologic Surgeons scale score (P < .001), and Glasgow Coma Scale score (P = .003) and worse outcomes (modified Rankin Scale at discharge; P = .001). There was no significant association between NSE level and Fisher grade (P = .81) in multivariable analysis. CONCLUSION: We found a significant association between higher NSE levels and poorer clinical presentations and worse outcomes. Although it is still early for any relevant clinical conclusions, our results suggest that NSE holds promise as a tool for screening patients at increased risk of poor outcomes after SAH.


Subject(s)
Hemorrhage/blood , Phosphopyruvate Hydratase/blood , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/therapy , Adult , Aged , Aged, 80 and over , Female , Glasgow Coma Scale , Humans , Isophane Insulin, Human , Male , Middle Aged , Predictive Value of Tests , Prognosis , Retrospective Studies , Subarachnoid Hemorrhage/enzymology , Treatment Outcome , Young Adult
17.
J Vasc Res ; 52(2): 103-15, 2015.
Article in English | MEDLINE | ID: mdl-26184661

ABSTRACT

Vasospasm is known to contribute to delayed cerebral ischemia following subarachnoid hemorrhage (SAH). We hypothesized that vasospasm initiates structural changes within the vessel wall, possibly aggravating ischemia and leading to resistance to vasodilator treatment. We therefore investigated the effect of blood on cerebral arteries with respect to contractile activation and vascular remodeling. In vitro experiments on rodent basilar and middle cerebral arteries showed a gradual contraction in response to overnight exposure to blood. After incubation with blood, a clear inward remodeling was found, reducing the caliber of the passive vessel. The transglutaminase inhibitor L682.777 fully prevented this remodeling. Translation of the in vitro findings to an in vivo SAH model was attempted in rats, using both a single prechiasmatic blood injection model and a double cisterna magna injection model, and in mice, using a single prechiasmatic blood injection. However, we found no substantial changes in active or passive biomechanical properties in vivo. We conclude that extravascular blood can induce matrix remodeling in cerebral arteries, which reduces vascular caliber. This remodeling depends on transglutaminase activity. However, the current rodent SAH models do not permit in vivo confirmation of this mechanism.


Subject(s)
Middle Cerebral Artery/physiopathology , Subarachnoid Hemorrhage/physiopathology , Vascular Remodeling , Vasospasm, Intracranial/physiopathology , Animals , Biomechanical Phenomena , Blood Flow Velocity , Cerebrovascular Circulation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/enzymology , Middle Cerebral Artery/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Rats, Wistar , Regional Blood Flow , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology , Transglutaminases/antagonists & inhibitors , Transglutaminases/genetics , Transglutaminases/metabolism , Vascular Remodeling/drug effects , Vasoconstriction , Vasospasm, Intracranial/enzymology , Vasospasm, Intracranial/genetics , Vasospasm, Intracranial/pathology
18.
Int J Mol Sci ; 16(6): 14171-80, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26110388

ABSTRACT

Delayed cerebral vasospasm is an important pathological feature of subarachnoid hemorrhage (SAH). The cause of vasospasm is multifactorial. Impairs nitric oxide availability and endothelial nitric oxide synthase (eNOS) dysfunction has been reported to underlie vasospasm. Memantine, a low-affinity uncompetitive N-methyl-d-aspartate (NMDA) blocker has been proven to reduce early brain injury after SAH. This study investigated the effect of memantine on attenuation of vasospasm and restoring eNOS functionality. Male Sprague-Dawley rats weighing 350-450 g were randomly divided into three weight-matched groups, sham surgery, SAH + vehicle, and SAH + memantine groups. The effects of memantine on SAH were evaluated by assessing the severity of vasospasm and the expression of eNOS. Memantine effectively ameliorated cerebral vasospasm by restoring eNOS functionality. Memantine can prevent vasospasm in experimental SAH. Treatment strategies may help combat SAH-induced vasospasm in the future.


Subject(s)
Disease Models, Animal , Endothelium, Vascular/drug effects , Memantine/pharmacology , Nitric Oxide Synthase Type II/metabolism , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/drug therapy , Animals , Blotting, Western , Endothelium, Vascular/enzymology , Excitatory Amino Acid Antagonists/pharmacology , Male , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/enzymology , Vasospasm, Intracranial/enzymology , Vasospasm, Intracranial/etiology
19.
J Clin Invest ; 125(7): 2609-25, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26011640

ABSTRACT

Subarachnoid hemorrhage (SAH) carries a 50% mortality rate. The extravasated erythrocytes that surround the brain contain heme, which, when released from damaged red blood cells, functions as a potent danger molecule that induces sterile tissue injury and organ dysfunction. Free heme is metabolized by heme oxygenase (HO), resulting in the generation of carbon monoxide (CO), a bioactive gas with potent immunomodulatory capabilities. Here, using a murine model of SAH, we demonstrated that expression of the inducible HO isoform (HO-1, encoded by Hmox1) in microglia is necessary to attenuate neuronal cell death, vasospasm, impaired cognitive function, and clearance of cerebral blood burden. Initiation of CO inhalation after SAH rescued the absence of microglial HO-1 and reduced injury by enhancing erythrophagocytosis. Evaluation of correlative human data revealed that patients with SAH have markedly higher HO-1 activity in cerebrospinal fluid (CSF) compared with that in patients with unruptured cerebral aneurysms. Furthermore, cisternal hematoma volume correlated with HO-1 activity and cytokine expression in the CSF of these patients. Collectively, we found that microglial HO-1 and the generation of CO are essential for effective elimination of blood and heme after SAH that otherwise leads to neuronal injury and cognitive dysfunction. Administration of CO may have potential as a therapeutic modality in patients with ruptured cerebral aneurysms.


Subject(s)
Heme Oxygenase-1/physiology , Membrane Proteins/physiology , Microglia/enzymology , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/enzymology , Acute-Phase Reaction/cerebrospinal fluid , Animals , Apoptosis , Carbon Monoxide/administration & dosage , Carbon Monoxide/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Erythrocytes/pathology , Female , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/cerebrospinal fluid , Heme Oxygenase-1/deficiency , Humans , Intracranial Aneurysm/cerebrospinal fluid , Intracranial Aneurysm/enzymology , Male , Maze Learning/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Phagocytosis/physiology , Protoporphyrins/pharmacology , Subarachnoid Hemorrhage/pathology
20.
Stroke ; 46(7): 1916-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25991416

ABSTRACT

BACKGROUND AND PURPOSE: Acute communicating hydrocephalus and cerebral edema are common and serious complications of subarachnoid hemorrhage (SAH), whose causes are poorly understood. Using a mouse model of SAH, we determined whether soluble epoxide hydrolase (sEH) gene deletion protects against SAH-induced hydrocephalus and edema by increasing levels of vasoprotective eicosanoids and suppressing vascular inflammation. METHODS: SAH was induced via endovascular puncture in wild-type and sEH knockout mice. Hydrocephalus and tissue edema were assessed by T2-weighted magnetic resonance imaging. Endothelial activation was assessed in vivo using T2*-weighted magnetic resonance imaging after intravenous administration of iron oxide particles linked to anti-vascular cell adhesion molecule-1 antibody 24 hours after SAH. Behavioral outcome was assessed at 96 hours after SAH with the open field and accelerated rotarod tests. RESULTS: SAH induced an acute sustained communicating hydrocephalus within 6 hours of endovascular puncture in both wild-type and sEH knockout mice. This was followed by tissue edema, which peaked at 24 hours after SAH and was limited to white matter fiber tracts. sEH knockout mice had reduced edema, less vascular cell adhesion molecule-1 uptake, and improved outcome compared with wild-type mice. CONCLUSIONS: Genetic deletion of sEH reduces vascular inflammation and edema and improves outcome after SAH. sEH inhibition may serve as a novel therapy for SAH.


Subject(s)
Brain Edema/enzymology , Epoxide Hydrolases/deficiency , Subarachnoid Hemorrhage/enzymology , Vasculitis/enzymology , Animals , Brain Edema/pathology , Inflammation/enzymology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Subarachnoid Hemorrhage/pathology , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...