Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Front Cell Neurosci ; 15: 710481, 2021.
Article En | MEDLINE | ID: mdl-34646122

Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular disease with high mortality and disability. Aberrant neuroinflammation has been identified as a critical factor accounting for the poor prognosis of SAH patients. Mast cells (MCs), the sentinel cells of the immune system, play a critical in the early immune reactions and participate in multiple pathophysiological process. However, the exact role of MCs on the pathophysiological process after SAH has not been fully understood. The current study was conducted to determine the role of MCs and MC stabilization in the context of SAH. Mouse SAH model was established by endovascular perforation process. Mice received saline or cromolyn (MC stabilizer) or compound 48/80 (MCs degranulator). Post-SAH evaluation included neurobehavioral test, western blot, immunofluorescence, and toluidine blue staining. We demonstrated that SAH induced MCs activation/degranulation. Administration of MC stabilizer cromolyn conferred a better neurologic outcome and decreased brain edema when compared with SAH+vehicle group. Furthermore, cromolyn significantly inhibited neuroinflammatory response and alleviated neuronal damage after SAH. However, pharmacological activation of MCs with compound 48/80 dramatically aggravated SAH-induced brain injury and exacerbated neurologic outcomes. Notably, pharmacological inhibition of microglial PAR-2 significantly reversed MCs-induced inflammatory response and neurological impairment. Additionally, the effect of MCs-derived tryptase in mediating neuroinflammation was also abolished by the microglial PAR-2 blockage in vitro. Taken together, MCs yielded inflammatory injury through activating microglia-related neuroinflammation after SAH. These data shed light on the notion that MCs might be a novel and promising therapeutic target for SAH.

2.
J Neuroinflammation ; 18(1): 154, 2021 Jul 07.
Article En | MEDLINE | ID: mdl-34233703

BACKGROUND: Complex changes in the brain microenvironment following traumatic brain injury (TBI) can cause neurological impairments for which there are few efficacious therapeutic interventions. The reactivity of astrocytes is one of the keys to microenvironmental changes, such as neuroinflammation, but its role and the molecular mechanisms that underpin it remain unclear. METHODS: Male C57BL/6J mice were subjected to the controlled cortical impact (CCI) to develop a TBI model. The specific ligand of AXL receptor tyrosine kinase (AXL), recombinant mouse growth arrest-specific 6 (rmGas6) was intracerebroventricularly administered, and selective AXL antagonist R428 was intraperitoneally applied at 30 min post-modeling separately. Post-TBI assessments included neurobehavioral assessments, transmission electron microscopy, immunohistochemistry, and western blotting. Real-time polymerase chain reaction (RT-PCR), siRNA transfection, and flow cytometry were performed for mechanism assessments in primary cultured astrocytes. RESULTS: AXL is upregulated mainly in astrocytes after TBI and promotes astrocytes switching to a phenotype that exhibits the capability of ingesting degenerated neurons or debris. As a result, this astrocytic transformation promotes the limitation of neuroinflammation and recovery of neurological dysfunction. Pharmacological inhibition of AXL in astrocytes significantly decreased astrocytic phagocytosis both in vivo and in primary astrocyte cultures, in contrast to the effect of treatment with the rmGas6. AXL activates the signal transducer and activator of the transcription 1 (STAT1) pathway thereby further upregulating ATP-binding cassette transporter 1 (ABCA1). Moreover, the supernatant from GAS6-depleted BV2 cells induced limited enhancement of astrocytic phagocytosis in vitro. CONCLUSION: Our work establishes the role of AXL in the transformation of astrocytes to a phagocytic phenotype via the AXL/STAT1/ABCA1 pathway which contributes to the separation of healthy brain tissue from injury-induced cell debris, further ameliorating neuroinflammation and neurological impairments after TBI. Collectively, our findings provide a potential therapeutic target for TBI.


Astrocytes/enzymology , Brain Injuries, Traumatic/metabolism , Cerebral Cortex/enzymology , Phagocytosis/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/pathology , Cells, Cultured , Cerebral Cortex/pathology , Male , Mice , Mice, Inbred C57BL , Axl Receptor Tyrosine Kinase
3.
Aging (Albany NY) ; 13(12): 16816-16833, 2021 06 28.
Article En | MEDLINE | ID: mdl-34182541

BACKGROUND: Subarachnoid hemorrhage (SAH) is a life-threatening disease worldwide, and effective pharmaceutical treatment is still lacking. Celastrol is a plant-derived triterpene which showed neuroprotective potential in several types of brain insults. This study aimed to investigate the effects of celastrol on early brain injury (EBI) after SAH. METHODS: A total of sixty-one male Sprague-Dawley rats were used in this study. Rat SAH endovascular perforation model was established to mimic the pathological changes of EBI after SAH. Multiple methods such as 3.0T MRI scanning, immunohistochemistry, western blotting and propidium iodide (PI) labeling were used to explore the therapeutic effects of celastrol on SAH. RESULTS: Celastrol treatment attenuated SAH-caused brain swelling, reduced T2 lesion volume and ventricular volume in MRI scanning, and improved overall neurological score. Albumin leakage and the degradation of tight junction proteins were also ameliorated after celastrol administration. Celastrol protected blood-brain bairrer integrity through inhibiting MMP-9 expression and anti-neuroinflammatory effects. Additionally, necroptosis-related proteins RIP3 and MLKL were down-regulated and PI-positive cells in the basal cortex were less in the celastrol-treated SAH group than that in untreated SAH group. CONCLUSIONS: Celastrol exhibits neuroprotective effects on EBI after SAH and deserves to be further investigated as an add-on pharmaceutical therapy.


Blood-Brain Barrier/pathology , Brain Injuries/drug therapy , Brain Injuries/etiology , Necroptosis/drug effects , Neuroprotective Agents/therapeutic use , Pentacyclic Triterpenes/therapeutic use , Subarachnoid Hemorrhage/complications , Albumins/metabolism , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Brain Edema/complications , Brain Edema/diagnostic imaging , Brain Edema/drug therapy , Brain Injuries/diagnostic imaging , Cerebral Ventricles/drug effects , Cerebral Ventricles/pathology , Down-Regulation/drug effects , Inflammation/pathology , Male , Matrix Metalloproteinase 9/metabolism , Neuroprotective Agents/pharmacology , Organ Size/drug effects , Pentacyclic Triterpenes/pharmacology , Protein Kinases/metabolism , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Subarachnoid Hemorrhage/diagnostic imaging , Survival Analysis , Tight Junctions/drug effects , Tight Junctions/metabolism , Up-Regulation/drug effects
4.
Neurosci Bull ; 37(4): 535-549, 2021 Apr.
Article En | MEDLINE | ID: mdl-33421025

Ferroptosis is a form of iron-dependent regulated cell death. Evidence of its existence and the effects of its inhibitors on subarachnoid hemorrhage (SAH) is still lacking. In the present study, we found that liproxstatin-1 protected HT22 cells against hemin-induced injury by protecting mitochondrial functions and ameliorating lipid peroxidation. In in vivo experiments, we demonstrated the presence of characteristic shrunken mitochondria in ipsilateral cortical neurons after SAH. Moreover, liproxstatin-1 attenuated the neurological deficits and brain edema, reduced neuronal cell death, and restored the redox equilibrium after SAH. The inhibition of ferroptosis by liproxstatin-1 was associated with the preservation of glutathione peroxidase 4 and the downregulation of acyl-CoA synthetase long-chain family member 4 as well as cyclooxygenase 2. In addition, liproxstatin-1 decreased the activation of microglia and the release of IL-6, IL-1ß, and TNF-α. These data enhance our understanding of cell death after SAH and shed light on future preclinical studies.


Ferroptosis , Subarachnoid Hemorrhage , Animals , Quinoxalines , Rats , Rats, Sprague-Dawley , Spiro Compounds , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy
5.
World Neurosurg ; 146: e955-e960, 2021 02.
Article En | MEDLINE | ID: mdl-33217589

OBJECTIVE: The best management of asymptomatic moyamoya disease (MMD) remains controversial. In this study, the authors aimed to explore an experience for treatment modality for asymptomatic MMD. METHODS: The authors retrospectively reviewed a total of 23 patients (age range 30-58 years) with asymptomatic MMD during the past 5 years at their institutions. The patients were divided into 2 groups: The surgical group included 11 patients, and the conservative group included 12 patients. The demographic, radiologic, and clinical findings of the patients were evaluated. At follow-up over 13-65 months, the future clinical and radiologic progression events were evaluated. RESULTS: During the follow-up period, 3 patients suffered from future clinical progression events in the conservative group: 1 experienced stroke, and 2 experienced transient ischemic attack. Among the patients in the surgical group, only 1 experienced transient ischemic attack. Kaplan-Meier analysis showed that patients undergoing surgeries had longer clinical progression-free survival times compared with patients in the conservative group (P = 0.002). CONCLUSIONS: Surgical treatment may be an alternative choice for patients with asymptomatic MMD. However, the best strategy for asymptomatic MMD in order to reduce future cerebrovascular risks still needs to be further explored.


Cerebral Revascularization/methods , Conservative Treatment , Ischemic Attack, Transient/epidemiology , Moyamoya Disease/therapy , Stroke/epidemiology , Adult , Angiography, Digital Subtraction , Asymptomatic Diseases , Cerebral Angiography , Disease Progression , Female , Humans , Ischemic Attack, Transient/etiology , Magnetic Resonance Angiography , Male , Middle Aged , Moyamoya Disease/complications , Progression-Free Survival , Retrospective Studies , Stroke/etiology
6.
J Neuroinflammation ; 17(1): 165, 2020 May 25.
Article En | MEDLINE | ID: mdl-32450897

BACKGROUND: Neuroinflammation is closely associated with the poor prognosis in subarachnoid hemorrhage (SAH) patients. This study was aimed to determine the role of stimulator of IFN genes (STING), an essential regulator to innate immunity, in the context of SAH. METHODS: A total of 344 male C57BL/6 J mice were subjected to endovascular perforation to develop a model of SAH. Selective STING antagonist C-176 and STING agonist CMA were administered at 30 min or 1 h post-modeling separately. To investigate the underlying mechanism, the AMPK inhibitor compound C was administered intracerebroventricularly at 30 min before surgery. Post-SAH assessments included SAH grade, neurological test, brain water content, western blotting, RT-PCR, and immunofluorescence. Oxygenated hemoglobin was introduced into BV2 cells to establish a SAH model in vitro. RESULTS: STING was mainly distributed in microglia, and microglial STING expression was significantly increased after SAH. Administration of C-176 substantially attenuated SAH-induced brain edema and neuronal injury. More importantly, C-176 significantly alleviated both short-term and persistent neurological dysfunction after SAH. Meanwhile, STING agonist CMA remarkably exacerbated neuronal injury and deteriorated neurological impairments. Mechanically, STING activation aggravated neuroinflammation via promoting microglial activation and polarizing into M1 phenotype, evidenced by microglial morphological changes, as well as the increased level of microglial M1 markers including IL-1ß, iNOS, IL-6, TNF-α, MCP-1, and NLRP3 inflammasome, while C-176 conferred a robust anti-inflammatory effect. However, all the mentioned beneficial effects of C-176 including alleviated neuroinflammation, attenuated neuronal injury and the improved neurological function were reversed by AMPK inhibitor compound C. Meanwhile, the critical role of AMPK signal in C-176 mediated anti-inflammatory effect was also confirmed in vitro. CONCLUSION: Microglial STING yielded neuroinflammation after SAH, while pharmacologic inhibition of STING could attenuate SAH-induced inflammatory injury at least partly by activating AMPK signal. These data supported the notion that STING might be a potential therapeutic target for SAH.


Inflammation/pathology , Membrane Proteins/metabolism , Subarachnoid Hemorrhage/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Subarachnoid Hemorrhage/immunology , Subarachnoid Hemorrhage/metabolism
7.
Medicine (Baltimore) ; 98(46): e17670, 2019 Nov.
Article En | MEDLINE | ID: mdl-31725609

This study aims to identify predictive factors related to clinical outcome, reoperation, and complications in patients with brain abscess.Patients with a diagnosis of brain abscess at discharge at the Second Affiliated Hospital of Zhejiang University School of Medicine between 2008 and 2018 were reviewed. Logistic regression was used to identify predictive factors associated with Glasgow Outcome Scale (GOS) at discharge, GOS at 1 year after discharge, reoperation and complications.Among 183 patients enrolled into the study, 142 patients had a good outcome at discharge (GOS ≥ 4) and 41 had a poor outcome (GOS ≤ 3). During the follow-up period, 20 additional patients had a good outcome. A total of 156 patients were treated by open craniotomy excision (n = 72) and aspiration (n = 84), 10 of whom underwent reoperation. Complications in surgical patients for brain abscess occurred in 54 patients. Poor outcome was related to Glasgow coma scale (P = .007) and ventricular proximity (P = .001). Surgical method was associated with reoperation (P = .04) and complications (P < .001). Seizure at admission was related to epilepsy (P < .001). Surgical method was related to postoperative intracranial hemorrhage (P = .02).Glasgow coma scale (GCS) and ventricular proximity were associated with poor outcome. Further, patients who underwent aspiration were more likely to experience reoperation, while open craniotomy excision (OCE) was related to complications. Patients presenting seizure at admission were more likely to develop epilepsy. Patients who underwent OCE tended to experience postoperative intracranial hemorrhage.


Brain Abscess/surgery , Glasgow Outcome Scale , Intracranial Hemorrhages/etiology , Postoperative Complications/etiology , Reoperation/statistics & numerical data , Adolescent , Adult , Aged , Aged, 80 and over , Brain Abscess/pathology , Child , Child, Preschool , Craniotomy/adverse effects , Craniotomy/methods , Female , Glasgow Coma Scale , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome , Young Adult
8.
Cell Transplant ; 28(11): 1358-1372, 2019 11.
Article En | MEDLINE | ID: mdl-31370690

Despite the substantial efforts to elucidate the role of early brain injury in subarachnoid hemorrhage (SAH), an effective pharmaceutical therapy for patients with SAH continues to be unavailable. This study aims to reveal the role of necroptosis after SAH, and explore whether the disruption of the blood-brain barrier (BBB) and RIP3-mediated necroptosis following SAH in a rat SAH model are altered by necrostatin-1 via its selective inhibition of receptor-interacting protein kinase 1 (RIP1). Sixty-five rats were used in the experiments. The SAH model was established using endovascular perforation. Necrostatin-1 was intracerebroventricularly injected 1 h before SAH induction. The neuroprotective effects of necrostatin-1 were evaluated with multiple methods such as magnetic resonance imaging (MRI) scanning, immunohistochemistry, propidium iodide (PI) labeling, and western blotting. Pretreatment with necrostatin-1 attenuated brain swelling and reduced the lesion volume on T2 sequence and ventricular volume on MRI 72 h after SAH induction. Albumin leakage and the degradation of tight junction proteins were also ameliorated by necrostatin-1 administration. In addition, necrostatin-1 decreased the number of PI-positive cells in the basal cortex, reduced the levels of the RIP3 and MLKL proteins, and inhibited the production of the pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α. Based on the findings from the present study, the selective RIP1 inhibitor necrostatin-1 functioned as a neuroprotective agent after SAH by attenuating brain swelling and BBB disruption. Moreover, the necrostatin-1 pretreatment prevented SAH-induced necroptosis by suppressing the activity of the RIP3/MLKL signaling pathway. These results will provide insights into new drugs and pharmacological targets to manage SAH, which are worth further study.


Blood-Brain Barrier/metabolism , Brain/drug effects , Imidazoles/therapeutic use , Indoles/therapeutic use , Necroptosis/drug effects , Neuroprotective Agents/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Subarachnoid Hemorrhage/drug therapy , Animals , Brain/cytology , Brain/diagnostic imaging , Brain/pathology , Brain Edema/drug therapy , Cytokines/metabolism , Magnetic Resonance Imaging , Matrix Metalloproteinase 9/metabolism , Necroptosis/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/pathology , Tight Junctions/drug effects , Tight Junctions/pathology
9.
FASEB J ; 33(2): 1727-1741, 2019 02.
Article En | MEDLINE | ID: mdl-30211660

This study aimed to explore the neuroprotective effect of mesencephalic astrocyte-derived neurotrophic factor (MANF) protein on early brain injury caused by subarachnoid hemorrhage (SAH) and the relevant mechanisms in experimental rats, expecting to understand whether MANF was a potential therapeutic target for SAH treatment. A perforation model of SAH was introduced into the study. Recombinant human MANF (rh-MANF) and protein kinase B (Akt) inhibitor (MK2206) were used to explore the effect and the mechanisms. Multiple approaches for systemic assessment were employed in the research, including the Garcia test, the SAH grade, Evans blue (EB) dye leakage, brain-water content (BWC), the rotarod test, and the Morris water-navigation task, as were biotechniques, such as immunohistochemistry, Western blot, transmission electron microscopy, and flow cytometry. MANF was mainly expressed in rat neurons, and its expression increased significantly at 3 h after SAH induction and peaked at 24 h. Stereotactic injection of rh-MANF into the cerebroventricle significantly increased the level of MANF, p-Akt, p-mouse double minute 2 homolog (p-MDM2), and B-cell lymphoma 2 (Bcl-2) in brain tissue, whereas it down-regulated the expression of P53, Bcl-2-associated X protein (Bax), and cleaved caspase-3, which indicated that neuronal apoptosis was remarkably suppressed. Expression of matrix metallopeptidase 9 (MMP-9) was also suppressed by the rh-MANF injection. Furthermore, neurologic deficits, EB dye leakage, and BWC were reduced, and long-lasting neuroprotection was noted with rh-MANF administration. The antiapoptotic and blood-brain barrier (BBB) protective effect could be offset by administering MK2206. MANF could alleviate neuronal apoptosis by activating Akt-dependent prosurvival pathway and abate BBB damage via MMP-9 suppression. MANF showed not only transient but also long-lasting neuroprotective properties. The rh-MANF as a potential drug for treating SAH might be of clinical use.-Li, T., Xu, W., Gao, L., Guan, G., Zhang, Z., He, P., Xu, H., Fan, L., Yan, F., Chen, G. Mesencephalic astrocyte-derived neurotrophic factor affords neuroprotection to early brain injury induced by subarachnoid hemorrhage via activating Akt-dependent prosurvival pathway and defending blood-brain barrier integrity.


Blood-Brain Barrier , Brain Injuries/prevention & control , Nerve Growth Factors/physiology , Proto-Oncogene Proteins c-akt/metabolism , Subarachnoid Hemorrhage/complications , Animals , Brain Injuries/etiology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Line, Tumor , Humans , Male , Rats , Rats, Sprague-Dawley , Subcellular Fractions/pathology
10.
Phys Med Biol ; 64(3): 035009, 2019 01 24.
Article En | MEDLINE | ID: mdl-30566918

Small animals such as non-human primate (NHP) and rodent are valuable models in frontier neuroscience researches, and comparative research between the animal model and human is helpful to understand and reveal the functional brain circuits in cognition and underlying mechanism in psychological disease. Small animals can be trained or anesthetized to endure long-term and multiple imaging scans; however, the concomitant needs in subcortical structure and function investigations pose major challenges in, e.g. spatial resolution, scan time efficiency, spatial/temporal signal-to-noise-ratio, as well as apparatus mechanical fixation. In addition, comparative investigations across species are also expected to be conducted under similar physical environment (such as the main magnetic field strength, RF pulse shape, sequence protocols, gradient waveform, system stability, etc in MRI), in order to avoid possible deviation in signal detection under different platforms, as well as to reduce experiment complexity. We have proposed a novel 5-channel surface coil that is equipped on 7T human MRI scanners and designed for small animal structural and functional MRI. Through a series of in vivo experiments over an anesthetized rat and macaque, the presented coil shows its main characteristics in, i.e. flexible coil mounting, reduced FOV, high temporal SNR, and parallel imaging capability. Such design is able to compensate the relatively lower gradient slew rate of human scanners versus those with smaller bores, and thus effectively facilitates in vivo microscopic structural MR images being obtained within a shortened and safe period of anesthesia; besides, it also enables high-resolution functional MRI (i.e. spin-echo based) being achieved with reasonable temporal resolution, distortion level and functional contrast.


Magnetic Resonance Imaging/instrumentation , Animals , Female , Humans , Image Processing, Computer-Assisted , Macaca mulatta , Male , Phantoms, Imaging , Rats , Signal-To-Noise Ratio
11.
Am J Chin Med ; 46(6): 1225-1241, 2018.
Article En | MEDLINE | ID: mdl-30149758

Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disease with few effective pharmacotherapies available. Salvia miltiorrhiza, a traditional Chinese medicinal herb, has been widely used to treat cardiovascular diseases for centuries. Recent studies have demonstrated that magnesium lithospermate B (MLB), a bioactive ingredient extracted from Salvia miltiorrhiza, exerts neuroprotective effects in several central nervous system insults. However, little is known about the role of MLB in SAH-induced brain injury and the exact molecular mechanism. In the current study, we studied the neuroprotective effects of MLB in SAH and explored the potential mechanism. Adult male Sprague-Dawley rats were subjected to an endovascular perforation process to produce an SAH model. MLB was administrated intraperitoneally at 30[Formula: see text]min after SAH with a dose of 25[Formula: see text]mg/kg or 50[Formula: see text]mg/kg. We found that administration of MLB significantly attenuated brain edema and neurological deficits after SAH. In addition, immunofluorescence staining demonstrated that MLB dose-dependently inhibited the activation of microglia and reduced neuronal apoptosis. Western blot analysis showed that MLB decreased the expression of inflammatory cytokine TNF-[Formula: see text] and pro-apoptotic protein cleaved caspase-3. More importantly, MLB increased the expression of SIRT1, while inhibited the acetylation of NF-[Formula: see text]B. Furthermore, pretreatment with sirtinol (a selective inhibitor of SIRT1) reversed all the aforementioned effects of MLB after SAH. In conclusion, our results indicated that MLB exerted robust neuroprotective effects against SAH via suppressing neuroinflammation and apoptosis. These neuroprotective effects of MLB against SAH might be exerted via regulating the SIRT1/NF-[Formula: see text]B pathway. MLB or the SIRT1/NF-[Formula: see text]B pathway could be a novel and promising therapeutic strategy for SAH management.


Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Neuroprotective Agents , Phytotherapy , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis , Disease Models, Animal , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/isolation & purification , Inflammation Mediators/metabolism , Infusions, Parenteral , Male , Microglia/pathology , NF-kappa B/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Salvia miltiorrhiza/chemistry , Signal Transduction/drug effects , Sirtuin 1/metabolism , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/pathology , Tumor Necrosis Factor-alpha/metabolism
12.
Biomed Pharmacother ; 107: 563-570, 2018 Nov.
Article En | MEDLINE | ID: mdl-30114640

Necroptosis is an inflammatory form of cell death that depends on receptor-interacting serine-threonine kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) and displays the morphological characteristics of necrosis. To date, it is unclear to what extent necroptosis contributes to subarachnoid hemorrhage (SAH) induced brain injury. The present study aimed to investigate the RIPK3-mediated necroptosis and the effects of the RIPK3 selective inhibitor GSK'872 in early brain injury following SAH. After SAH, RIPK3 expression increased as early as 6 h and peaked at 72 h. Double immunofluorescence staining revealed that RIPK3 was mainly located in neurons. Most necrotic cells were neurons, which were further confirmed by TEM. Intracerebroventricular injection of GSK'872 (25 mM) could attenuate brain edema and improve neurological function following SAH and reduce the number of necrotic cells. In addition, GSK'872 could also decrease the protein levels of RIPK3 and MLKL, and cytoplasmic translocation and expression of HMGB1, an important pro-inflammatory protein. Taken together, the current study provides the new evidence that RIPK3-mediated necroptosis is involved in early brain injury and GSK'872 decreases the RIPK3-mediated necroptosis and subsequent cytoplasmic translocation and expression of HMGB1, as well as ameliorates brain edema and neurological deficits.


Apoptosis , Brain Injuries/etiology , Brain Injuries/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Subarachnoid Hemorrhage/complications , Animals , Apoptosis/drug effects , Cytoplasm/metabolism , HMGB1 Protein/metabolism , Male , Necrosis , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Transport/drug effects , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
13.
Thromb Res ; 167: 64-73, 2018 07.
Article En | MEDLINE | ID: mdl-29793135

OBJECTIVES: The non-specific clinical manifestations and lack of effective diagnostic techniques have made cerebral venous sinus thrombosis (CVST) difficult to recognize and easy to misdiagnose. Several studies have suggested that different types of magnetic resonance venography (MRV) have advantages in diagnosing CVST. We conducted this meta-analysis to assess the accuracy of MRV in identifying CVST. MATERIAL AND METHODS: We searched the Embase, PubMed, and Chinese Biomedical (CBM) databases comprehensively to retrieve eligible articles up to Mar 31, 2018. The methodological quality of each article was evaluated individually. The summary diagnostic accuracy of MRV for CVST was obtained from pooled analysis with random-effects models. Sensitivity analysis, subgroup analysis, and meta-regression were used to explore the sources of heterogeneity. A trim and fill analysis was conducted to correct the funnel plot asymmetry. RESULTS: The meta-analysis synthesized 12 articles containing 27 cohorts with a total of 1933 cases. The pooled sensitivity and specificity were 0.86 (95% CI: 0.83, 0.89) and 0.94 (95% CI: 0.93, 0.95), respectively. The pooled diagnostic odds ratio (DOR) was 75.24 (95% CI: 38.33, 147.72). The area under the curve (AUC) was 0.9472 (95% CI: 0.9229, 0.9715). Subgroup analysis and meta-regression analysis revealed the technical types of MRV and the methods of counting cases contributing to the heterogeneity. The trim and fill method confirmed that publication bias has little effect on our results. CONCLUSIONS: MRV has excellent diagnostic performance and is accurate in confirming CVST.


Magnetic Resonance Spectroscopy/methods , Phlebography/methods , Sinus Thrombosis, Intracranial/diagnostic imaging , Sinus Thrombosis, Intracranial/diagnosis , Humans , Sinus Thrombosis, Intracranial/pathology
14.
Biomed Pharmacother ; 99: 947-955, 2018 Mar.
Article En | MEDLINE | ID: mdl-29710495

Phosphodiesterase-4 (PDE4) plays a fundamental role in a range of central nervous system (CNS) insults, however, the role of PDE4 in early brain injury (EBI) after subarachnoid hemorrhage (SAH) remains unclear. The current study was designed to investigate the role of PDE4 in EBI after SAH and explore the potential mechanism. The SAH model in Sprague-Dawley rat was established by endovascular perforation process. Rats were randomly divided into: sham group, SAH?+?vehicle group, SAH?+?rolipram (PDE4 inhibitor) group, SAH?+?rolipram?+?sirtinol (SIRT1 inhibitor) group and SAH?+?rolipram+MK2206 (Akt inhibitor) group. Mortality, SAH grades, neurological function, brain edema, immunofluorescence staining and western blotting were performed. Double fluorescence labeling staining indicated that PDE4 was located predominately in neurons after SAH. Rolipram reduced brain edema, improved neurological function in the rat model of SAH. Moreover, rolipram increased the expression of Sirtuin1 (SIRT1) and up-regulated the phosphorylation of Akt, which was accompanied by the reduction of neuronal apoptosis. Administration of sirtinol inhibited the phosphorylation of Akt. Moreover, all the beneficial effects of rolipram against SAH were abolished by both sirtinol and MK2206. These data indicated that PDE4 inhibition by rolipram protected rats against EBI after SAH via suppressing neuronal apoptosis through the SIRT1/Akt pathway. Rolipram might be an important therapeutic drug for SAH.


Neuroprotective Agents/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Rolipram/pharmacology , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis/drug effects , Benzamides/administration & dosage , Benzamides/pharmacology , Brain Edema/prevention & control , Brain Injuries/prevention & control , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Male , Naphthols/administration & dosage , Naphthols/pharmacology , Neuroprotective Agents/administration & dosage , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Rolipram/administration & dosage , Sirtuin 1/metabolism , Subarachnoid Hemorrhage/pathology
15.
Neurochem Res ; 43(4): 785-795, 2018 Apr.
Article En | MEDLINE | ID: mdl-29397536

Early brain injury (EBI) is the primary cause of poor outcome in subarachnoid hemorrhage (SAH) patients. Rolipram, a specific phosphodiesterase-4 inhibitor which is traditionally used as an anti-depressant drug, has been recently proven to exert neuroprotective effects in several central nervous system insults. However, the role of rolipram in SAH remains uncertain. The current study was aimed to investigate the role of rolipram in EBI after SAH and explore the potential mechanism. Adult male Sprague-Dawley rats were subjected to an endovascular perforation process to produce an SAH model. Rolipram was injected intraperitoneally at 2 h after SAH with a dose of 10 mg/kg. We found that rolipram significantly ameliorated brain edema and alleviated neurological dysfunction after SAH. Rolipram treatment remarkably promoted the expression of Sirtuin 1 (SIRT1) while inhibited NF-κB activation. Moreover, rolipram significantly inhibited the activation of microglia as well as down-regulated the expression of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6. In addition, rolipram increased the expression of protective cytokine IL-10. Furthermore, rolipram significantly alleviated neuronal death after SAH. In conclusion, these data suggested that rolipram exerts neuroprotective effects against EBI after SAH via suppressing neuroinflammation and reducing neuronal loss. The neuroprotective effects of rolipram were associated with regulating the SIRT1/NF-κB pathway. Rolipram could be a novel and promising therapeutic agent for SAH treatment.


Brain Injuries/prevention & control , NF-kappa B/antagonists & inhibitors , Rolipram/administration & dosage , Sirtuin 1/biosynthesis , Subarachnoid Hemorrhage/drug therapy , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Injections, Intraperitoneal , Male , NF-kappa B/metabolism , Neuroprotective Agents/administration & dosage , Phosphodiesterase 4 Inhibitors/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology
16.
Oncotarget ; 8(43): 73547-73558, 2017 Sep 26.
Article En | MEDLINE | ID: mdl-29088725

BACKGROUND: Hydrogen sulfide (H2S) has shown a neuroprotective role in several cerebrovascular diseases. This study aimed to explore the underlying mechanisms of H2S in early brain injury after subarachnoid hemorrhage (SAH). METHODS: One hundred seventy-seven male Sprague-Dawley rats were employed in this study. Sodium hydrosulfide (NaHS), a donor of H2S, was injected intraperitoneally at 60 min after SAH was induced by endovascular perforation. Western blot analysis determined the expression of several proteins of interest, and an immunofluorescence assay was used to examine neuronal apoptosis. RESULTS: Exogenous NaHS markedly improved neurological scores, attenuated brain edema, and ameliorated neuronal apoptosis at 24 h after SAH induction. The underlying mechanisms of H2S in ameliorating neuronal apoptosis might be executed through inhibition of the activity of mammalian sterile 20-like kinase 1 (MST1) protein. Western blot analysis demonstrated that exogenous NaHS decreased cleaved MST1 (cl-MST1) while increasing full-length MST1 expression. This anti-apoptotic effect of H2S could be reversed by chelerythrine, which could activate MST1 via caspase-dependent cleavage. CONCLUSIONS: Exogenous NaHS, as a donor of H2S, could ameliorate early brain injury after SAH by inhibiting neuronal apoptosis by reducing the activity of the MST1 protein.

17.
J Neuroinflammation ; 14(1): 186, 2017 Sep 13.
Article En | MEDLINE | ID: mdl-28903766

BACKGROUND: The NLRP3 inflammasome is a multiprotein complex that regulates the innate immune inflammatory response by activating caspase-1 and subsequent IL-1ß and IL-18. Fluoxetine has been shown to have the anti-inflammatory properties in many disease models. However, the effects and mechanisms of these effects of fluoxetine in early brain injury after subarachnoid hemorrhage (SAH) have not been defined. METHODS: The SAH model was induced by an endovascular perforation in adult male Sprague-Dawley (SD) rats weighing 300-320 g. N-Ac-Tyr-Val-Ala-Asp-chloromethyl ketone (AC-YVAD-CMK) was injected intraperitoneally (5 mg/kg) 1 h after SAH. Fluoxetine was administered via intravenous route 6 h after SAH. 3-Methyladenine (3-MA) was intracerebroventricularly injected 20 min before SAH. SAH grade, neurological function, brain water content, propidium iodide (PI) staining, western blot, double immunostaining, and transmission electron microscopy were performed. RESULTS: Expression of caspase-1 increased and peaked at 24 h after SAH. Caspase activation was along with the increased necrotic cells, which occurred mainly in neurons. Necrotic cell death of microglia and astrocyte were also found. Administration of AC-YVAD-CMK, a caspase-1 inhibitor, reduced the expression of IL-1ß and IL-18 and the number of PI-positive cells, attenuated brain edema, and improved neurological function, which was also observed in fluoxetine-treated rats. Furthermore, fluoxetine treatment significantly decreased the expression of NLRP3 and cleaved caspase-1 and upregulated the expression of beclin-1, a marker for autophagy. Finally, the effects of fluoxetine in NLRP3 inflammasome activation were reversed by additional 3-MA administration. CONCLUSIONS: Together, our present study indicated that NLRP3 inflammasome and caspase-1 activation play a deleterious role in early brain injury and fluoxetine mitigates NLRP3 inflammasome and caspase-1 activation through autophagy activation after SAH, providing a potential therapeutic agent for SAH treatment.


Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Fluoxetine/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Subarachnoid Hemorrhage/pathology , Animals , Brain Injuries/immunology , Brain Injuries/metabolism , Brain Injuries/pathology , Inflammasomes/drug effects , Inflammasomes/immunology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/immunology , Subarachnoid Hemorrhage/metabolism
18.
Free Radic Biol Med ; 112: 336-349, 2017 11.
Article En | MEDLINE | ID: mdl-28790012

Aberrant modulation of mitochondrial dynamic network, which shifts the balance of fusion and fission towards fission, is involved in brain damage of various neurodegenerative diseases including Parkinson's disease, Huntington's disease and Alzheimer's disease. A recent research has shown that the inhibition of mitochondrial fission alleviates early brain injury after experimental subarachnoid hemorrhage, however, the underlying molecular mechanisms have remained to be elucidated. This study was undertaken to characterize the effects of the inhibition of dynamin-related protein-1 (Drp1, a dominator of mitochondrial fission) on blood-brain barrier (BBB) disruption and neuronal apoptosis following SAH and the potential mechanisms. The endovascular perforation model of SAH was performed in adult male Sprague Dawley rats. The results indicated Mdivi-1(a selective Drp1 inhibitor) reversed the morphologic changes of mitochondria and Drp1 translocation, reduced ROS levels, ameliorated the BBB disruption and brain edema remarkably, decreased the expression of MMP-9 and prevented degradation of tight junction proteins-occludin, claudin-5 and ZO-1. Mdivi-1 administration also inhibited the nuclear translocation of nuclear factor-kappa B (NF-κB), leading to decreased expressions of TNF-ɑ, IL-6 and IL-1ß. Moreover, Mdivi-1 treatment attenuated neuronal cell death and improved neurological outcome. To investigate the underlying mechanisms further, we determined that Mdivi-1 reduced p-PERK, p-eIF2α, CHOP, cleaved caspase-3 and Bax expression as well as increased Bcl-2 expression. Rotenone (a selective inhibitor of mitochondrial complexes I) abolished both the anti-BBB disruption and anti-apoptosis effects of Mdivi-1. In conclusion, these data implied that excessive mitochondrial fission might inhibit mitochondrial complex I to become a cause of oxidative stress in SAH, and the inhibition of Drp1 by Mdivi-1 attenuated early brain injury after SAH probably via the suppression of inflammation-related blood-brain barrier disruption and endoplasmic reticulum stress-based apoptosis.


Blood-Brain Barrier/drug effects , Dynamins/genetics , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Quinazolinones/pharmacology , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Dynamins/antagonists & inhibitors , Dynamins/metabolism , Endoplasmic Reticulum Stress/drug effects , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Occludin/genetics , Occludin/metabolism , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/pathology , Subarachnoid Space/drug effects , Subarachnoid Space/metabolism , Subarachnoid Space/pathology , Survival Analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
19.
Brain Behav Immun ; 65: 125-139, 2017 Oct.
Article En | MEDLINE | ID: mdl-28457811

Subarachnoid hemorrhage (SAH) is a serious medical problem with few effective pharmacotherapies available, and neuroinflammation has been identified as an important pathological process in early brain injury (EBI) after SAH. Methylene blue (MB) is an older drug that has been recently proven to exert extraordinary neuroprotective effects in several brain insults. However, no study has reported the beneficial effects of MB in SAH. In the current investigation, we studied the neuroprotective effects of MB in EBI after SAH and focused on its anti-inflammatory role. A total of 303 rats were subjected to an endovascular perforation process to produce an SAH model. We found that MB could significantly ameliorate brain edema secondary to BBB disruption and alleviate neurological dysfunction after SAH. MB administration also promoted the phosphorylation of Akt and GSK-3ß, leading to an increased concentration of MEF2D in the nucleus. The cytokine IL-10 was up-regulated, and IL-1ß, IL-6 and TNF-α were down-regulated after MB administration. MB administration could also alleviate neutrophil infiltration and microglia activation after SAH. MK2206, a selective inhibitor of Akt, abolished the neuroprotective effects of MB, inhibited the phosphorylation of Akt and prevented the nuclear localization of MEF2D. MK2206 also reduced the expression of IL-10 and increased the expression of pro-inflammatory cytokines. In conclusion, these data suggested that MB could ameliorate neuroinflammatory responses after SAH, and its anti-inflammatory effects might be exerted via activation of the Akt/GSK-3ß/MEF2D pathway.


Methylene Blue/pharmacology , Subarachnoid Hemorrhage/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/drug effects , Brain/metabolism , Brain Edema/drug therapy , Brain Injuries/metabolism , Cytokines/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , MEF2 Transcription Factors/metabolism , Male , Neuroimmunomodulation/drug effects , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/physiopathology , Tumor Necrosis Factor-alpha/metabolism
20.
Neural Plast ; 2017: 5405104, 2017.
Article En | MEDLINE | ID: mdl-28255460

Traumatic brain injury (TBI) is a serious medical and social problem worldwide. Because of the complex pathophysiological mechanisms of TBI, effective pharmacotherapy is still lacking. The microglial cells are resident tissue macrophages located in the brain and have two major polarization states, M1 phenotype and M2 phenotype, when activated. The M1 phenotype is related to the release of proinflammatory cytokines and secondary brain injury, while the M2 phenotype has been proved to be responsible for the release of anti-inflammation cytokines and for central nervous system (CNS) repair. In animal models, pharmacological strategies inhibiting the M1 phenotype and promoting the M2 phenotype of microglial cells could alleviate cerebral damage and improve neurological function recovery after TBI. In this review, we aimed to summarize the current knowledge about the pathological significance of microglial M1/M2 polarization in the pathophysiology of TBI. In addition, we reviewed several drugs that have provided neuroprotective effects against brain injury following TBI by altering the polarization states of the microglia. We emphasized that future investigation of the regulation mechanisms of microglial M1/M2 polarization in TBI is anticipated, which could contribute to the development of new targets of pharmacological intervention in TBI.


Brain Injuries, Traumatic/metabolism , Brain/drug effects , Brain/metabolism , Encephalitis/metabolism , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Cell- and Tissue-Based Therapy/methods , Cytokines/metabolism , Encephalitis/complications , Humans , Inflammation Mediators , Neuroprotective Agents/therapeutic use
...