Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.186
1.
CNS Neurosci Ther ; 30(3): e14681, 2024 03.
Article En | MEDLINE | ID: mdl-38516845

BACKGROUND: Peroxiredoxin 2 (Prx2), an intracellular protein that regulates redox reactions, released from red blood cells is involved in inflammatory brain injury after intracerebral hemorrhage (ICH). Toll-like receptor 4 (TLR4) may be crucial in this process. This study investigated the role of the Prx2-TLR4 inflammatory axis in brain injury following experimental ICH in mice. METHODS: First, C57BL/6 mice received an intracaudate injection of autologous arterial blood or saline and their brains were harvested on day 1 to measure Prx2 levels. Second, mice received an intracaudate injection of either recombinant mouse Prx2 or saline. Third, the mice were co-injected with autologous arterial blood and conoidin A, a Prx2 inhibitor, or vehicle. Fourth, the mice received a Prx2 injection and were treated with TAK-242, a TLR4 antagonist, or saline (intraperitoneally). Behavioral tests, magnetic resonance imaging, western blot, immunohistochemistry/immunofluorescence staining, and RNA sequencing (RNA-seq) were performed. RESULTS: Brain Prx2 levels were elevated after autologous arterial blood injection. Intracaudate injection of Prx2 caused brain swelling, microglial activation, neutrophil infiltration, neuronal death, and neurological deficits. Co-injection of conoidin A attenuated autologous arterial blood-induced brain injury. TLR4 was expressed on the surface of microglia/macrophages and neutrophils and participated in Prx2-induced inflammation. TAK-242 treatment attenuated Prx2-induced inflammation and neurological deficits. CONCLUSIONS: Prx2 can cause brain injury following ICH through the TLR4 pathway, revealing the Prx2-TLR4 inflammatory axis as a potential therapeutic target.


Brain Injuries , Sulfonamides , Toll-Like Receptor 4 , Animals , Mice , Brain Injuries/etiology , Cerebral Hemorrhage/metabolism , Inflammation/etiology , Inflammation/pathology , Mice, Inbred C57BL , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Peroxiredoxins/therapeutic use , Toll-Like Receptor 4/metabolism
3.
Methods Mol Biol ; 2761: 569-588, 2024.
Article En | MEDLINE | ID: mdl-38427262

Traumatic brain injury (TBI) is one of the foremost causes of disability and death globally. Prerequisites for successful therapy of disabilities associated with TBI involved improved knowledge of the neurobiology of TBI, measurement of quantitative changes in recovery dynamics brought about by therapy, and the translation of quantitative methodologies and techniques that were successful in tracking recovery in preclinical models to human TBI. Frequently used animal models of TBI in research and development include controlled cortical impact, fluid percussion injury, blast injury, penetrating blast brain injury, and weight-drop impact acceleration models. Preclinical models of TBI benefit from controlled injury settings and the best prospects for biometric quantification of injury and therapy-induced gradual recovery from disabilities. Impact acceleration closed head TBI paradigm causes diffuse TBI (DTBI) without substantial focal brain lesions in rats. DTBI is linked to a significant rate of death, morbidity, and long-term disability. DTBI is difficult to diagnose at the time of hospitalization with imaging techniques making it challenging to take prompt therapeutic action. The weight-drop method without craniotomy is an impact acceleration closed head DTBI model that is used to induce mild/moderate diffuse brain injuries in rodents. Additionally, we have characterized neuropathological and neurobehavioral outcomes of the weight-drop model without craniotomy for inducing closed head DTBI of graded severity with a range of mass of weights (50-450 gm). This chapter also discusses techniques and protocols for measuring numerous functional disabilities and pathological changes in the brain brought on by DTBI.


Blast Injuries , Brain Injuries, Traumatic , Brain Injuries , Humans , Rats , Animals , Disease Models, Animal , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/complications , Brain Injuries/etiology , Craniotomy
4.
J Stroke Cerebrovasc Dis ; 33(5): 107670, 2024 May.
Article En | MEDLINE | ID: mdl-38438086

BACKGROUND AND PURPOSE: The pathophysiological mechanisms underlying brain injury resulting from intracerebral hemorrhage (ICH) remain incompletely elucidated, and efficacious therapeutic interventions to enhance the prognosis of ICH patients are currently lacking. Previous research indicates that MicroRNA-7 (miR-7) can suppress the expression of Nod-like receptor protein 3 (NLRP3), thereby modulating neuroinflammation in Parkinson's disease pathogenesis. However, the potential regulatory effects miR-7 on NLRP3 inflammasome after ICH are yet to be established. This study aims to ascertain whether miR-7 mitigates secondary brain injury following experimental ICH by inhibiting NLRP3 and to investigate the underlying mechanisms. METHODS: An ICH model was established by stereotaxically injecting 100 µL of autologous blood into the right basal ganglia of Sprague-Dawley (SD) rats. Subsequently, these rats were allocated into three groups: sham, ICH + Vehicle, and ICH + miR-7, each comprising 18 animals. Twelve hours post-modeling, rats received intraventricular injections of 10 µL physiological saline, 10 µL phosphate, and 10 µL phosphate-buffered saline solution containing 0.5 nmol of miR-7 mimics, respectively. Neurological function was assessed on day three post-modeling, followed by euthanasia for brain tissue collection. Brain water content was determined using the dry-wet weight method. The expression of inflammatory cytokines in cerebral tissues surrounding the hematoma was analyzed through immunohistochemistry and Western blot assays. These cytokines were re-evaluated using Reverse Transcription-Polymerase Chain Reaction (RT-PCR). Moreover, bioinformatics tools were employed to predict miR-7's binding to NLRP3. A wild-type luciferase reporter gene vector and a corresponding mutant vector were constructed, followed by transfection of miR-7 mimics into HEK293T cells to assess luciferase activity. RESULTS: Our study demonstrates that the administration of miR-7 mimics markedly reduced neurological function scores and attenuated brain edema in rats following ICH. A significant upregulation of NLRP3 expression in microglia/macrophage adjacent to the hematoma was observed, substantially reduced after the treatment with miR-7 mimics. Furthermore, this intervention ameliorated neurodegenerative changes and effectively decreased the protein and mRNA levels of pro-inflammatory cytokines, namely TNF-α, IL-1ß, IL-6, and Caspase1, in the cerebral tissues proximate to the hematomas. In addition, miR-7 mimics distinctly inhibited the luciferase activity associated with the wild-type reporter gene, an effect not mirrored in its mutant variant. CONCLUSIONS: The miR-7 suppressed NLRP3 expression in microglia/macrophage to reduce the production of inflammatory cytokines, leading to conducting certain neuroprotection post-ICH in rats.


Brain Injuries , MicroRNAs , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Rats , Brain Injuries/etiology , Cerebral Hemorrhage/complications , Cytokines/genetics , Cytokines/metabolism , HEK293 Cells , Hematoma/complications , Luciferases/therapeutic use , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphates , Rats, Sprague-Dawley
5.
Exp Neurol ; 375: 114731, 2024 May.
Article En | MEDLINE | ID: mdl-38373483

The utilization of explosives and chemicals has resulted in a rise in blast-induced traumatic brain injury (bTBI) in recent times. However, there is a dearth of diagnostic biomarkers and therapeutic targets for bTBI due to a limited understanding of biological mechanisms, particularly in the early stages. The objective of this study was to examine the early neuropathological characteristics and underlying biological mechanisms of primary bTBI. A total of 83 Sprague Dawley rats were employed, with their heads subjected to a blast shockwave of peak overpressure ranging from 172 to 421 kPa in the GI, GII, and GIII groups within a closed shock tube, while the body was shielded. Neuromotor dysfunctions, morphological changes, and neuropathological alterations were detected through modified neurologic severity scores, brain water content analysis, MRI scans, histological, TUNEL, and caspase-3 immunohistochemical staining. In addition, label-free quantitative (LFQ)-proteomics was utilized to investigate the biological mechanisms associated with the observed neuropathology. Notably, no evident damage was discernible in the GII and GI groups, whereas mild brain injury was observed in the GIII group. Neuropathological features of bTBI were characterized by morphologic changes, including neuronal injury and apoptosis, cerebral edema, and cerebrovascular injury in the shockwave's path. Subsequently, 3153 proteins were identified and quantified in the GIII group, with subsequent enriched neurological responses consistent with pathological findings. Further analysis revealed that signaling pathways such as relaxin signaling, hippo signaling, gap junction, chemokine signaling, and sphingolipid signaling, as well as hub proteins including Prkacb, Adcy5, and various G-protein subunits (Gnai2, Gnai3, Gnao1, Gnb1, Gnb2, Gnb4, and Gnb5), were closely associated with the observed neuropathology. The expression of hub proteins was confirmed via Western blotting. Accordingly, this study proposes signaling pathways and key proteins that exhibit sensitivity to brain injury and are correlated with the early pathologies of bTBI. Furthermore, it highlights the significance of G-protein subunits in bTBI pathophysiology, thereby establishing a theoretical foundation for early diagnosis and treatment strategies for primary bTBI.


Blast Injuries , Brain Injuries, Traumatic , Brain Injuries , Rats , Animals , Protein Subunits , Blast Injuries/complications , Blast Injuries/pathology , Rats, Sprague-Dawley , Brain Injuries, Traumatic/metabolism , Brain Injuries/diagnostic imaging , Brain Injuries/etiology
7.
Neurotherapeutics ; 21(2): e00326, 2024 Mar.
Article En | MEDLINE | ID: mdl-38301326

Intraventricular hemorrhage (IVH) is an important cause of long-term disability in extremely preterm infants, with no current treatment. This study assessed the potential neuroprotective effects of cannabidiol (CBD) in an IVH model using immature rats. IVH was induced in 1-day-old (P1) Wistar rats by left periventricular injection of Clostridial collagenase. Some rats received CBD prenatally (10 â€‹mg/kg i.p. to the dam) and then 5 â€‹mg/kg i.p. 6, 30 and 54 â€‹h after IVH (IVH+CBD, n â€‹= â€‹30). Other IVH rats received vehicle (IVH+VEH, n â€‹= â€‹34) and vehicle-treated non-IVH rats served as controls (SHM, n â€‹= â€‹29). Rats were humanely killed at P6, P14 or P45. Brain damage (motor and memory performance, area of damage, Lactate/N-acetylaspartate ratio), white matter injury (ipsilateral hemisphere and corpus callosum volume, oligodendroglial cell density and myelin basic protein signal), blood-brain barrier (BBB) integrity (Mfsd2a, occludin and MMP9 expression, gadolinium leakage), inflammation (TLR4, NFκB and TNFα expression, infiltration of pro-inflammatory cells), excitotoxicity (Glutamate/N-acetylspartate ratio) and oxidative stress (protein nitrosylation) were then evaluated. CBD prevented the long-lasting motor and cognitive consequences of IVH, reduced brain damage in the short- and long-term, protected oligodendroglial cells preserving adequate myelination and maintained BBB integrity. The protective effects of CBD were associated with the modulation of inflammation, excitotoxicity and oxidative stress. In conclusion, in immature rats, CBD reduced IVH-induced brain damage and its short- and long-term consequences, showing robust and pleiotropic neuroprotective effects. CBD is a potential candidate to ameliorate IVH-induced immature brain damage.


Brain Injuries , Cannabidiol , Neuroprotective Agents , Humans , Infant, Newborn , Animals , Rats , Blood-Brain Barrier , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Infant, Premature/metabolism , Rats, Wistar , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Brain Injuries/drug therapy , Brain Injuries/etiology , Inflammation , Disease Models, Animal
8.
Exp Neurol ; 373: 114679, 2024 Mar.
Article En | MEDLINE | ID: mdl-38190933

INTRODUCTION: We studied spatiotemporal features of acute transcriptional inflammatory response induced by a focal brain injury in distant uninjured neuronal tissue and a role of endocannabinoid (eCB) system in its control. MATERIALS AND METHODS: A focal excitotoxic lesion was induced by a unilateral injection of kainate in the dorsal hippocampus of awake Wistar rats. During acute post-injury period (3 h and 24 h post-injection), mRNA levels of genes associated with neuroinflammation (Il1b, Il6, Tnf, Ccl2; Cx3cl1, Zc3 h12a, Tgfb1) and eCB receptors of CB1 and CB2 types (Cnr1 and Cnr2) in intact regions of the hippocampus and neocortex were measured using qPCR. Occurrence of acute symptomatic seizures was controlled electrographically. To modulate eCB signaling during injury and acute post-injury period, antagonists (AM251, AM630) and agonist (WIN55-212-2) of eCB receptors were administered before the injury induction. RESULTS: Local intrahippocampal injury triggered widespread time- and region-dependent neuroinflammation in undamaged brain regions remote from the lesion site. The distant areas of the hippocampus and hippocampal meninges exhibited early (3 h) transient upregulation of pro- and anti-inflammatory cytokines simultaneously with occurrence of acute symptomatic seizures. The neocortex and its meninges showed minor neuroinflammation early after injury (3 h) but later (24 h) significantly upregulated several genes, mainly with anti-inflammatory properties. Focal lesion also changed expression of eCB receptors in the distant extra-lesional regions - CB1 receptors at 3 h and both CB1 and CB2 receptors at 24 h. Within the hippocampus, significant regional differences in constitutive and post-injury expression CB1 receptors were found. Pharmacological blockade of eCB receptors during injury and early post-injury period lengthened hippocampal neuroinflammation and reversed upregulation of anti-inflammatory molecules in the neocortex. CONCLUSION: The findings show that focal brain injury rapidly triggers widespread parenchymal and extraparenchymal neuroinflammation. The early injury-induced response is likely to represent neurogenic neuroinflammation produced by network hyperexcitability (acute symptomatic seizures). Activation of eCB signaling during acute phase of the brain injury is important for initiation of adaptive anti-inflammatory processes and prevention of chronic pathologic neuroinflammation in distant uninjured structures. However, the beneficial role of injury-induced eCB activity appears to depend on many factors including time, brain region, eCB tone etc.


Brain Injuries , Endocannabinoids , Rats , Animals , Endocannabinoids/metabolism , Rats, Wistar , Neuroinflammatory Diseases , Hippocampus/metabolism , Seizures , Brain Injuries/etiology , Anti-Inflammatory Agents , Receptor, Cannabinoid, CB1/metabolism
9.
Transl Stroke Res ; 15(1): 219-237, 2024 02.
Article En | MEDLINE | ID: mdl-36631632

Subarachnoid hemorrhage (SAH) is a type of stroke with high morbidity and mortality. Netrin-1 (NTN-1) can alleviate early brain injury (EBI) following SAH by enhancing peroxisome proliferator-activated receptor gamma (PPARγ), which is an important transcriptional factor modulating lipid metabolism. Ferroptosis is a newly discovered type of cell death related to lipid metabolism. However, the specific function of ferroptosis in NTN-1-mediated neuroprotection following SAH is still unclear. This study aimed to evaluate the neuroprotective effects and the possible molecular basis of NTN-1 in SAH-induced EBI by modulating neuronal ferroptosis using the filament perforations model of SAH in mice and the hemin-stimulated neuron injury model in HT22 cells. NTN-1 or a vehicle was administered 2 h following SAH. We examined neuronal death, brain water content, neurological score, and mortality. NTN-1 treatment led to elevated survival probability, greater survival of neurons, and increased neurological score, indicating that NTN-1-inhibited ferroptosis ameliorated neuron death in vivo/in vitro in response to SAH. Furthermore, NTN-1 treatment enhanced the expression of PPARγ, nuclear factor erythroid 2-related factor 2 (Nrf2), and glutathione peroxidase 4 (GPX4), which are essential regulators of ferroptosis in EBI after SAH. The findings show that NTN-1 improves neurological outcomes in mice and protects neurons from death caused by neuronal ferroptosis. Furthermore, the mechanism underlying NTN-1 neuroprotection is correlated with the inhibition of ferroptosis, attenuating cell death via the PPARγ/Nrf2/GPX4 pathway and coenzyme Q10-ferroptosis suppressor protein 1 (CoQ10-FSP1) pathway.


Brain Injuries , Ferroptosis , Subarachnoid Hemorrhage , Rats , Mice , Animals , NF-E2-Related Factor 2/metabolism , PPAR gamma , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Netrin-1/pharmacology , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/metabolism , Signal Transduction
10.
Transl Stroke Res ; 15(2): 476-494, 2024 04.
Article En | MEDLINE | ID: mdl-36781743

Early brain injury (EBI) is the leading cause of poor prognosis for patients suffering from subarachnoid hemorrhage (SAH), particularly learning and memory deficits in the repair phase. A recent report has involved calcium/calmodulin-dependent protein kinase II (CaMKII) in the pathophysiological process underlying SAH-induced EBI. Alpha-asarone (ASA), a major compound isolated from the Chinese medicinal herb Acorus tatarinowii Schott, was proven to reduce secondary brain injury by decreasing CaMKII over-phosphorylation in rats' model of intracerebral hemorrhage in our previous report. However, the effect of ASA on SAH remains unclear, and the role of CaMKII in both acute and recovery stages of SAH needs further investigation. In this work, we first established a classic SAH rat model by endovascular perforation and intraperitoneally administrated different ASA doses (10, 20, and 40 mg/kg) 2 h after successful modeling. Then, the short- and long-term neurobehavioral performances were blindly evaluated to confirm ASA's efficacy against SAH. Subsequently, we explored ASA's therapeutic mechanism in both acute and recovery stages using histopathological examination, TUNEL staining, flow cytometry, Western-blot, double-immunofluorescence staining, and transmission electron microscopy (TEM) observation. Finally, KN93, a selective CaMKII inhibitor, was applied in oxyhemoglobin-damaged HT22 cells to explore the role of CaMKII in ASA's neuroprotective effect. The results demonstrated that ASA alleviated short- and long-term neurological dysfunction, reduced mortality and seizure rate within 24 h, and prolonged 14-day survival in SAH rats. Histopathological examination showed a reduction of neuronal damage and a restoration of the hippocampal structure after ASA treatment in both acute and recovery phases of SAH. In the acute stage, the Western-blot and flow cytometer analyses showed that ASA restored E/I balance, reduced calcium overload and CaMKII phosphorylation, and inhibited mitochondrion-involved apoptosis, thus preventing neuronal damage and apoptosis underlying EBI post-SAH. In the recovery stage, the TEM observation, double-immunofluorescence staining, and Western-blot analyses indicated that ASA increased the numbers of synapses and enhanced synaptic plasticity in the ipsilateral hippocampi, probably by promoting NR2B/CaMKII interaction and activating subsequent CREB/BDNF/TrkB signaling pathways. Furthermore, KN93 notably reversed ASA's neuroprotective effect on oxyhemoglobin-damaged HT22 cells, confirming CaMKII a potential target for ASA's efficacy against SAH. Our study confirmed for the first time that ASA ameliorated the SAH rats' neurobehavioral deterioration, possibly via modulating CaMKII-involved pathways. These findings provided a promising candidate for the clinical treatment of SAH and shed light on future drug discovery against SAH.


Allylbenzene Derivatives , Anisoles , Benzenesulfonamides , Benzylamines , Brain Injuries , Neuroprotective Agents , Subarachnoid Hemorrhage , Humans , Rats , Animals , Rats, Sprague-Dawley , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/pathology , Calcium/therapeutic use , Oxyhemoglobins/therapeutic use , Brain Injuries/etiology
11.
ASAIO J ; 70(3): 167-176, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38051987

Extracorporeal membrane oxygenation (ECMO) is a form of temporary cardiopulmonary bypass for patients with acute respiratory or cardiac failure refractory to conventional therapy. Its usage has become increasingly widespread and while reported survival after ECMO has increased in the past 25 years, the incidence of neurological injury has not declined, leading to the pressing question of how to improve time-to-detection and diagnosis of neurological injury. The neurological status of patients on ECMO is clinically difficult to evaluate due to multiple factors including illness, sedation, and pharmacological paralysis. Thus, increasing attention has been focused on developing tools and techniques to measure and monitor the brain of ECMO patients to identify dynamic risk factors and monitor patients' neurophysiological state as a function in time. Such tools may guide neuroprotective interventions and thus prevent or mitigate brain injury. Current means to continuously monitor and prevent neurological injury in ECMO patients are rather limited; most techniques provide indirect or postinsult recognition of irreversible brain injury. This review will explore the indications, advantages, and disadvantages of standard-of-care, emerging, and investigational technologies for neurological monitoring on ECMO, focusing on bedside techniques that provide continuous assessment of neurological health.


Brain Injuries , Extracorporeal Membrane Oxygenation , Heart Failure , Respiratory Insufficiency , Adult , Humans , Child , Extracorporeal Membrane Oxygenation/adverse effects , Extracorporeal Membrane Oxygenation/methods , Heart Failure/etiology , Brain , Brain Injuries/prevention & control , Brain Injuries/etiology , Respiratory Insufficiency/therapy , Retrospective Studies
12.
CNS Neurosci Ther ; 30(3): e14452, 2024 03.
Article En | MEDLINE | ID: mdl-37735980

AIMS: Rasd1 has been reported to be correlated with neurotoxicity, metabolism, and rhythm, but its effect in case of subarachnoid hemorrhage (SAH) remained unclear. White matter injury (WMI) and ferroptosis participate in the early brain injury (EBI) after SAH. In this work, we have investigated whether Rasd1 can cause ferroptosis and contribute to SAH-induced WMI. METHODS: Lentivirus for Rasd1 knockdown/overexpression was administrated by intracerebroventricular (i.c.v) injection at 7 days before SAH induction. SAH grade, brain water content, short- and long-term neurobehavior, Western blot, real-time PCR, ELISA, biochemical estimation, immunofluorescence, diffusion tensor imaging (DTI), and transmission electron microscopy (TEM) were systematically performed. Additionally, genipin, a selective uncoupling protein 2(UCP2) inhibitor, was used in primary neuron and oligodendrocyte co-cultures for further in vitro mechanistic studies. RESULTS: Rasd1 knockdown has improved the neurobehavior, glia polarization, oxidative stress, neuroinflammation, ferroptosis, and demyelination. Conversely, Rasd1 overexpression aggravated these changes by elevating the levels of reactive oxygen species (ROS), inflammatory cytokines, MDA, free iron, and NCOA4, as well as contributing to the decrease of the levels of UCP2, GPX4, ferritin, and GSH mechanistically. According to the in vitro study, Rasd1 can induce oligodendrocyte ferroptosis through inhibiting UCP2, increasing reactive oxygen species (ROS), and activating NCOA4-mediated ferritinophagy. CONCLUSIONS: It can be concluded that Rasd1 exerts a modulated role in oligodendrocytes ferroptosis in WMI following SAH.


Brain Injuries , Subarachnoid Hemorrhage , White Matter , Animals , Brain Injuries/etiology , Diffusion Tensor Imaging , Neurons/metabolism , Reactive Oxygen Species , Subarachnoid Hemorrhage/diagnostic imaging , Subarachnoid Hemorrhage/metabolism , White Matter/diagnostic imaging , White Matter/metabolism
13.
Neurocrit Care ; 40(2): 759-768, 2024 Apr.
Article En | MEDLINE | ID: mdl-37697125

BACKGROUND: Extracorporeal membrane oxygenation (ECMO) provides lifesaving support to critically ill patients who experience refractory cardiopulmonary failure but carries a high risk for acute brain injury. We aimed to identify characteristics reflecting acute brain injury in children requiring ECMO support. METHODS: This is a prospective observational study from 2019 to 2022 of pediatric ECMO patients undergoing neuromonitoring, including continuous electroencephalography, cerebral oximetry, and transcranial Doppler ultrasound (TCD). The primary outcome was acute brain injury. Clinical and neuromonitoring characteristics were collected. Multivariate logistic regression was implemented to model odds ratios (ORs) and identify the combined characteristics that best discriminate risk of acute brain injury using the area under the receiver operating characteristic curve. RESULTS: Seventy-five pediatric patients requiring ECMO support were enrolled in this study, and 62 underwent neuroimaging or autopsy evaluations. Of these 62 patients, 19 experienced acute brain injury (30.6%), including seven (36.8%) with arterial ischemic stroke, four (21.1%) with hemorrhagic stroke, seven with hypoxic-ischemic brain injury (36.8%), and one (5.3%) with both arterial ischemic stroke and hypoxic-ischemic brain injury. A univariate analysis demonstrated acute brain injury to be associated with maximum hourly seizure burden (p = 0.021), electroencephalographic suppression percentage (p = 0.022), increased interhemispheric differences in electroencephalographic total power (p = 0.023) and amplitude (p = 0.017), and increased differences in TCD Thrombolysis in Brain Ischemia (TIBI) scores between bilateral middle cerebral arteries (p = 0.023). Best subset model selection identified increased seizure burden (OR = 2.07, partial R2 = 0.48, p = 0.013), increased quantitative electroencephalographic interhemispheric amplitude differences (OR = 2.41, partial R2 = 0.48, p = 0.013), and increased interhemispheric TCD TIBI score differences (OR = 4.66, partial R2 = 0.49, p = 0.006) to be independently associated with acute brain injury (area under the receiver operating characteristic curve = 0.92). CONCLUSIONS: Increased seizure burden and increased interhemispheric differences in both quantitative electroencephalographic amplitude and TCD MCA TIBI scores are independently associated with acute brain injury in children undergoing ECMO support.


Brain Injuries , Brain Ischemia , Extracorporeal Membrane Oxygenation , Ischemic Stroke , Humans , Child , Extracorporeal Membrane Oxygenation/adverse effects , Extracorporeal Membrane Oxygenation/methods , Cerebrovascular Circulation/physiology , Oximetry , Brain Injuries/etiology , Brain Injuries/therapy , Middle Cerebral Artery , Seizures , Retrospective Studies
14.
Radiother Oncol ; 190: 109974, 2024 Jan.
Article En | MEDLINE | ID: mdl-37913956

BACKGROUND AND PURPOSE: Radiation-induced brain injury (RBI) is a severe radiotoxicity for nasopharyngeal carcinoma (NPC) patients, greatly affecting their long-term life quality and survival. We aim to establish a comprehensive predictive model including clinical factors and newly developed genetic variants to improve the precision of RBI risk stratification. MATERIALS AND METHODS: By performing a large registry-based retrospective study with magnetic resonance imaging follow-up on RBI development, we conducted a genome-wide association study and developed a polygenic risk score (PRS) for RBI in 1189 NPC patients who underwent intensity-modulated radiotherapy. We proposed a tolerance dose scheme for temporal lobe radiation based on the risk predicted by PRS. Additionally, we established a nomogram by combining PRS and clinical factors for RBI risk prediction. RESULTS: The 38-SNP PRS could effectively identify high-risk individuals of RBI (P = 1.42 × 10-34). Based on genetic risk calculation, the recommended tolerance doses of temporal lobes should be 57.6 Gy for individuals in the top 10 % PRS subgroup and 68.1 Gy for individuals in the bottom 50 % PRS. Notably, individuals with high genetic risk (PRS > P50) and receiving high radiation dose in the temporal lobes (D0.5CC > 65 Gy) had an approximate 50-fold risk over individuals with low PRS and receiving low radiation dose (HR = 50.09, 95 %CI = 24.27-103.35), showing an additive joint effect (Pinteraction < 0.001). By combining PRS with clinical factors including age, tumor stage, and radiation dose of temporal lobes, the predictive accuracy was significantly improved with C-index increased from 0.78 to 0.85 (P = 1.63 × 10-2). CONCLUSIONS: The PRS, together with clinical factors, could improve RBI risk stratification and implies personalized radiotherapy.


Brain Injuries , Nasopharyngeal Neoplasms , Radiotherapy, Intensity-Modulated , Humans , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Carcinoma/pathology , Retrospective Studies , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Nasopharyngeal Neoplasms/drug therapy , Genome-Wide Association Study , Brain Injuries/etiology , Radiotherapy, Intensity-Modulated/adverse effects , Risk Assessment
15.
J Pharm Pharmacol ; 76(1): 44-56, 2024 Jan 06.
Article En | MEDLINE | ID: mdl-37991718

OBJECTIVES: To investigate the protect effect of moslosooflavone against brain injury induced by hypobaric hypoxia (HH) in mice. METHODS: Protective effects of moslosooflavone in oxidative stress, neuroinflammation, energy metabolism disorder, and apoptosis were studied in HH-induced brain damage mice. The pathological morphology in the cortex of mice was determined by hematoxylin and eosin staining. The related protein expressions were detected by western blot. KEY FINDINGS: Moslosooflavone improved HH-induced brain histopathological changes, reduced the contents of ROS and MDA, and elevated the levels of antioxidant enzymes and GSH in HH-exposed brains of mice. Moslosooflavone also markedly enhanced the ATPase activities and PK, ATP contents, while reducing LDH activity and the LD, TNF-α, IL-1ß, and IL-6 contents HH-exposed brains of mice. In addition, moslosooflavone notably decreased the expression of HIF-1α, VEGF, Bax, and cleaved caspase-3 dramatically increasing the expression of Bcl-2, Nrf2, and HO­1 in HH-exposed brains of mice. CONCLUSIONS: Our current studies indicate that moslosooflavone protects HH-induced brain injury possibly through alleviating oxidative stress and neuroinflammation, maintaining the balance of energy metabolism, and inhibiting cell apoptosis.


Brain Injuries , Neuroinflammatory Diseases , Mice , Animals , Oxidative Stress , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/prevention & control , Hypoxia , Apoptosis , Energy Metabolism
16.
J Pediatr ; 266: 113838, 2024 Mar.
Article En | MEDLINE | ID: mdl-37995930

OBJECTIVE: To examine the relationship between perioperative brain injury and neurodevelopment during early childhood in patients with severe congenital heart disease (CHD). STUDY DESIGN: One hundred and seventy children with CHD and born at term who required cardiopulmonary bypass surgery in the first 6 weeks after birth were recruited from 3 European centers and underwent preoperative and postoperative brain MRIs. Uniform description of imaging findings was performed and an overall brain injury score was created, based on the sum of the worst preoperative or postoperative brain injury subscores. Motor and cognitive outcomes were assessed with the Bayley Scales of Infant and Toddler Development Third Edition at 12 to 30 months of age. The relationship between brain injury score and clinical outcome was assessed using multiple linear regression analysis, adjusting for CHD severity, length of hospital stay (LOS), socioeconomic status (SES), and age at follow-up. RESULTS: Neither the overall brain injury score nor any of the brain injury subscores correlated with motor or cognitive outcome. The number of preoperative white matter lesions was significantly associated with gross motor outcome after correction for multiple testing (P = .013, ß = -0.50). SES was independently associated with cognitive outcome (P < .001, ß = 0.26), and LOS with motor outcome (P < .001, ß = -0.35). CONCLUSION: Preoperative white matter lesions appear to be the most predictive MRI marker for adverse early childhood gross motor outcome in this large European cohort of infants with severe CHD. LOS as a marker of disease severity, and SES influence outcome and future intervention trials need to address these risk factors.


Brain Injuries , Heart Defects, Congenital , Infant , Humans , Child, Preschool , Brain/pathology , Brain Injuries/etiology , Brain Injuries/pathology , Heart Defects, Congenital/surgery , Heart Defects, Congenital/complications , Magnetic Resonance Imaging , Risk Factors
17.
Neuroreport ; 35(2): 90-97, 2024 Feb 07.
Article En | MEDLINE | ID: mdl-38109375

OBJECTIVES: Radiation therapy in the treatment of brain tumors also leads to the occurrence of radiation brain injury (RBI). Anlotinib is a small-molecule inhibitor of multi-receptor tyrosine kinase with high selectivity for vascular endothelial growth factor receptor-2. In this study, we constructed a rat model of RBI and investigated the effect of anlotinib on RBI and its mechanism of action through drug intervention during the acute phase of RBI. METHODS: Six-week-old male (Sprague-Dawley) rats were used to construct an animal model of RBI to evaluate the protective effect of anlotinib on acute RBI by histopathological staining, brain edema determination, blood-brain barrier integrity evaluation and quick real time-polymerase chain reaction , ELISA detection of inflammation-related indexes, and western-blot detection of related gene protein expression. RESULTS: Anlotinib reduced the degree of edema in the hippocampal region of rats, improved the pathological morphology of neural cells and vascular endothelial cells, and decreased blood-brain barrier permeability. Anlotinib reduced glial fibrillary acidic protein protein expression in the hippocampal region of rat brain tissue and inhibited astrocyte activation. It inhibited the release of inflammatory factors (interleukin [IL]-6, IL-8 and vascular endothelial growth factor) and down-regulated the expression of janus kinase-2/signal transducer and activator of transcription-3 (JAK2/STAT3) signaling pathway-related proteins. CONCLUSION: This study found that anlotinib has a protective effect against RBI in rats and anlotinib may be a new candidate for the treatment of RBI.


Brain Injuries , Endothelial Cells , Rats , Male , Animals , Rats, Sprague-Dawley , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Brain/metabolism , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism
18.
Cell Mol Biol (Noisy-le-grand) ; 69(12): 76-82, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-38063114

In recent years, numerous studies have demonstrated that tea polyphenols (TPPs) can exert neuroprotective effects through the regulation of the PI3K/Akt pathway. The objective of this work was to verify whether TPPs could protect against early brain injury in rats after subarachnoid hemorrhage (SAH) by modulating the PI3K/Akt pathway. A total of 150 rats were randomly rolled into control (C), TPP, and SAH groups. The TPP and SAH groups underwent endovascular perforation to induce SAH, while C group received only endovascular needle puncture and saline injection. Brain water content, Evans Blue (EB) extravasation assay, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, Western blot, and RT-PCR analyses were performed. Relative to SAH group, TPP treatment considerably improved neurological function scores following SAH, reduced brain edema, cortical neuronal apoptosis, and blood-brain barrier damage. Levels of aquaporin-4 (AQP4) and apoptosis-related protein Bax were considerably lower in the TPP group than in SAH group. Conversely, levels of anti-apoptotic protein Bcl-2 and tight junction protein Zona occludens 1 (ZO-1) were considerably higher in the TPP group. Furthermore, TPP treatment was found to activate the PI3K/Akt signaling. TPPs can mitigate early brain injury caused by SAH in rats by reducing AQP4 levels, alleviating cortical damage, and attenuating neuronal apoptosis. These findings elucidate the protective mechanisms of TPPs against early brain injury following SAH through the regulation of the PI3K/Akt signaling.


Brain Injuries , Neuroprotective Agents , Polyphenols , Subarachnoid Hemorrhage , Animals , Rats , Apoptosis , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Polyphenols/pharmacology , Polyphenols/therapeutic use
19.
Undersea Hyperb Med ; 50(4): 421-424, 2023.
Article En | MEDLINE | ID: mdl-38055883

Introduction: Cerebral radiation necrosis is rarely encountered in pediatric patients. This case report describes a child with cerebral radiation necrosis who was successfully treated using corticosteroids, bevacizumab, and hyperbaric oxygenation. Case report: A 3-year-old boy developed progressive extremity weakness six months after the completion of radiation therapy for the treatment of a neuroepithelial malignancy. Treatment with corticosteroids and bevacizumab was initiated, but his symptoms did not improve, and he was then referred for hyperbaric oxygen therapy. After completing 60 hyperbaric treatments, he experienced significant improvements in mobility, which remained stable over the next year. Discussion: Cerebral radiation necrosis typically presents in children with symptoms of ataxia or headache. Corticosteroids and bevacizumab are common treatments, but hyperbaric oxygen therapy has also been studied as a therapeutic modality for this condition. When considering the use of hyperbaric oxygenation in pediatric patients, careful attention to treatment planning and patient safety can reduce the risks of adverse events such as middle ear barotrauma and confinement anxiety. Conclusion: In addition to other available pharmacologic therapies, hyperbaric oxygenation should be considered for the treatment of pediatric patients with cerebral radiation necrosis.


Brain Injuries , Cerebrum , Hyperbaric Oxygenation , Radiation Injuries , Child, Preschool , Humans , Male , Barotrauma/etiology , Barotrauma/prevention & control , Bevacizumab/therapeutic use , Hyperbaric Oxygenation/adverse effects , Hyperbaric Oxygenation/methods , Necrosis/etiology , Necrosis/therapy , Cerebrum/pathology , Cerebrum/radiation effects , Brain Injuries/etiology , Brain Injuries/pathology , Brain Injuries/therapy , Radiation Injuries/etiology , Radiation Injuries/pathology , Radiation Injuries/therapy , Neoplasms, Neuroepithelial/radiotherapy
20.
Eur J Med Res ; 28(1): 479, 2023 Nov 04.
Article En | MEDLINE | ID: mdl-37925438

BACKGROUND: Older patients (aged ≥ 60 years) with severe brain injury have a high mortality and disability rate. The objective of this retrospective study was to assess the clinical risk factors of prognosis in older patients with severe brain injury after surgical intervention and to analyze the prognosis of the surviving group of patients 1 year after discharge. METHODS: Clinical data of older patients with severe brain injury who were admitted to two neurosurgical centers between January 2010 and December 2020 were collected. Patient age, sex, Glasgow Coma Scale (GCS) score at admission, underlying disease, mechanisms of injury, abnormal pupillary reflex, head computed tomography imaging findings (such as hematoma type),intraoperative brain swelling and other factors were reviewed. All the patients were categorized into a good prognosis (survival) group and a poor prognosis (death) group by the Glasgow Outcome Score (GOS); also, the related factors affecting the prognosis were screened and the independent risk factors were identified by the Binary logistic regression analysis. GOS was used to evaluate the prognosis of the surviving group of patients 1 year after discharge. RESULTS: Out of 269 patients, 171 (63.6%) survived, and 98 (36.4%) died during hospitalization. Univariate analysis showed that age, GCS score at admission, underlying diseases, abnormal pupillary reflex, the disappearance of ambient cistern, the midline structure shift, intraoperative brain swelling, oxygen saturation < 90%, and cerebral hernia were risk factors for the prognosis of older patients with severe brain injury after surgical intervention. Multivariate analysis showed that age, underlying diseases, disappearance of ambient cistern, Oxygen saturation < 90% and intraoperative brain swelling were independent risk factors of the prognosis in the population. The effect of surgical intervention differed among various age groups at 1-year follow-up after surgery. CONCLUSIONS: The results of this retrospective study confirmed that age, underlying diseases, disappearance of ambient cistern, intraoperative brain swelling, and oxygen saturation < 90% are associated with poor prognosis in older postoperative patients with severe brain injury. Surgical intervention may improve prognosis and reduce mortality in older patients (age < 75 years). But for those patients (age ≥ 75 years), the prognosis was poor after surgical intervention.


Brain Edema , Brain Injuries , Humans , Aged , Retrospective Studies , Prognosis , Risk Factors , Brain Injuries/etiology , Treatment Outcome
...