Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.683
Filter
1.
Front Immunol ; 15: 1386780, 2024.
Article in English | MEDLINE | ID: mdl-38756773

ABSTRACT

Introduction: Intracerebral hemorrhage (ICH) often triggers oxidative stress through reactive oxygen species (ROS). Transforming growth factor-ß-activated kinase 1 (TAK1) plays a pivotal role in regulating oxidative stress and inflammation across various diseases. 5Z-7-Oxozeaenol (OZ), a specific inhibitor of TAK1, has exhibited therapeutic effects in various conditions. However, the impact of OZ following ICH and its underlying molecular mechanisms remain elusive. This study aimed to explore the possible role of OZ in ICH and its underlying mechanisms by inhibiting oxidative stress-mediated pyroptosis. Methods: Adult male Sprague-Dawley rats were subjected to an ICH model, followed by treatment with OZ. Neurobehavioral function, blood-brain barrier integrity, neuronal pyroptosis, and oxidative stress markers were assessed using various techniques including behavioral tests, immunofluorescence staining, western blotting, transmission electron microscopy, and biochemical assays. Results: Our study revealed that OZ administration significantly inhibited phosphorylated TAK1 expression post-ICH. Furthermore, TAK1 blockade by OZ attenuated blood-brain barrier (BBB) disruption, neuroinflammation, and oxidative damage while enhancing neurobehavioral function. Mechanistically, OZ administration markedly reduced ROS production and oxidative stress by facilitating nuclear factor-erythroid 2-related factor 2 (NRF2) nuclear translocation. This was accompanied by a subsequent suppression of the NOD-like receptor protein 3 (NLRP3) activation-mediated inflammatory cascade and neuronal pyroptosis. Discussion: Our findings highlight that OZ alleviates brain injury and oxidative stress-mediated pyroptosis via the NRF2 pathway. Inhibition of TAK1 emerges as a promising approach for managing ICH.


Subject(s)
Cerebral Hemorrhage , MAP Kinase Kinase Kinases , NF-E2-Related Factor 2 , Neurons , Oxidative Stress , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Animals , Pyroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Male , Rats , Signal Transduction/drug effects , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Disease Models, Animal , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/drug therapy , Reactive Oxygen Species/metabolism , Lactones , Resorcinols , Zearalenone/administration & dosage
2.
Am J Chin Med ; 52(3): 799-819, 2024.
Article in English | MEDLINE | ID: mdl-38752843

ABSTRACT

Subarachnoid hemorrhage (SAH), a specific subtype of cerebrovascular accident, is characterized by the extravasation of blood into the interstice between the brain and its enveloping delicate tissues. This pathophysiological phenomenon can precipitate an early brain injury (EBI), which is characterized by inflammation and neuronal death. Rutaecarpine (Rut), a flavonoid compound discovered in various plants, has been shown to have protective effects against SAH-induced cerebral insult in rodent models. In our study, we used a rodent SAH model to evaluate the effect of Rut on EBI and investigated the effect of Rut on the inflammatory response and its regulation of SIRT6 expression in vitro. We found that Rut exerts a protective effect on EBI in SAH rats, which is partly due to its ability to inhibit the inflammatory response. Notably, Rut up-regulated Sirtuin 6 (SIRT6) expression, leading to an increase in H3K9 deacetylation and inhibition of nuclear factor-kappa B (NF-[Formula: see text]B) transcriptional activation, thereby mediating the inflammatory response. In addition, further data showed that SIRT6 was proven to mediate the regulation of Rut on the microglial inflammatory response. These findings highlight the importance of SIRT6 in the regulation of inflammation and suggest a potential mechanism for the protective effect of Rut on EBI. In summary, Rut may have the potential to prevent and treat SAH-induced brain injury by interacting with SIRT6. Our findings may provide a new therapeutic strategy for the treatment of SAH-induced EBI.


Subject(s)
Indole Alkaloids , NF-kappa B , Quinazolines , Rats, Sprague-Dawley , Sirtuins , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/complications , Sirtuins/metabolism , Sirtuins/genetics , Indole Alkaloids/pharmacology , NF-kappa B/metabolism , Male , Quinazolines/pharmacology , Quinazolines/therapeutic use , Disease Models, Animal , Brain Injuries/etiology , Brain Injuries/drug therapy , Brain Injuries/metabolism , Rats , Inflammation/drug therapy , Inflammation/etiology , Phytotherapy , Signal Transduction/drug effects , Gene Expression/drug effects , Quinazolinones
3.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 220-225, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814212

ABSTRACT

This study explored the impact of penehyclidine hydrochloride on cognitive function in rats with brain injury. Sprague-Dawley rats (n=36) were randomly assigned to sham-operation, model, and penehyclidine hydrochloride groups. Rats in the sham-operation group underwent craniotomy, while the model and penehyclidine hydrochloride groups received brain injury models and interventions with normal saline and penehyclidine hydrochloride, respectively. Specimens were obtained two weeks post-intervention. Neurological deficits were evaluated using Zea-Longa scores, and memory was assessed with the Morris water maze test. ELISA determined brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) content. mRNA expressions of BDNF and NGF were assessed via qPCR, and phosphorylated CREB (p-CREB) protein expression was measured by Western blotting. Compared to the sham-operation group, both model and penehyclidine hydrochloride groups showed increased Zea-Longa scores. Escape latencies were longer and platform crossings were fewer in model and penehyclidine hydrochloride groups compared to the sham-operation group, but penehyclidine hydrochloride demonstrated a shorter latency and more platform crossings than the model group. BDNF and NGF content decreased in model and penehyclidine hydrochloride groups compared to the sham-operation group, with an increase in the penehyclidine hydrochloride group compared to the model group. mRNA expression levels declined in model and penehyclidine hydrochloride groups but were higher in the latter. p-CREB protein expression was lower in model and penehyclidine hydrochloride groups compared to the sham-operation group but higher in the penehyclidine hydrochloride group than the model group. Penehyclidine hydrochloride exhibited neuroprotective effects by upregulating the cAMP/CREB signaling pathway, improving cognitive function in rats with brain injury.


Subject(s)
Brain Injuries , Brain-Derived Neurotrophic Factor , Cognition , Cyclic AMP Response Element-Binding Protein , Cyclic AMP , Quinuclidines , Rats, Sprague-Dawley , Signal Transduction , Animals , Signal Transduction/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Quinuclidines/pharmacology , Quinuclidines/therapeutic use , Cognition/drug effects , Male , Brain Injuries/drug therapy , Brain Injuries/metabolism , Cyclic AMP/metabolism , Rats , Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Phosphorylation/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Disease Models, Animal
4.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702778

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH), a severe subtype of stroke, is characterized by notably high mortality and morbidity, largely due to the lack of effective therapeutic options. Although the neuroprotective potential of PPARg and Nrf2 has been recognized, investigative efforts into oroxin A (OA), remain limited in preclinical studies. METHODS: SAH was modeled in vivo through filament perforation in male C57BL/6 mice and in vitro by exposing HT22 cells to hemin to induce neuronal damage. Following the administration of OA, a series of methods were employed to assess neurological behaviors, brain water content, neuronal damage, cell ferroptosis, and the extent of neuroinflammation. RESULTS: The findings indicated that OA treatment markedly improved survival rates, enhanced neurological functions, mitigated neuronal death and brain edema, and attenuated the inflammatory response. These effects of OA were linked to the suppression of microglial activation. Moreover, OA administration was found to diminish ferroptosis in neuronal cells, a critical factor in early brain injury (EBI) following SAH. Further mechanistic investigations uncovered that OA facilitated the translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus, thereby activating the Nrf2/GPX4 pathway. Importantly, OA also upregulated the expression of FSP1, suggesting a significant and parallel protective effect against ferroptosis in EBI following SAH in synergy with GPX4. CONCLUSION: In summary, this research indicated that the PPARg activator OA augmented the neurological results in rodent models and diminished neuronal death. This neuroprotection was achieved primarily by suppressing neuronal ferroptosis. The underlying mechanism was associated with the alleviation of cellular death through the Nrf2/GPX4 and FSP1/CoQ10 pathways.


Subject(s)
Ferroptosis , Mice, Inbred C57BL , Neuroinflammatory Diseases , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , Ferroptosis/drug effects , Ferroptosis/physiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/metabolism , Neurons/drug effects , Neurons/pathology
5.
Cell Mol Neurobiol ; 44(1): 36, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637346

ABSTRACT

Surgical brain injury (SBI), induced by neurosurgical procedures or instruments, has not attracted adequate attention. The pathophysiological process of SBI remains sparse compared to that of other central nervous system diseases thus far. Therefore, novel and effective therapies for SBI are urgently needed. In this study, we found that neutrophil extracellular traps (NETs) were present in the circulation and brain tissues of rats after SBI, which promoted neuroinflammation, cerebral edema, neuronal cell death, and aggravated neurological dysfunction. Inhibition of NETs formation by peptidylarginine deiminase (PAD) inhibitor or disruption of NETs with deoxyribonuclease I (DNase I) attenuated SBI-induced damages and improved the recovery of neurological function. We show that SBI triggered the activation of cyclic guanosine monophosphate-adenosine monophosphate synthase stimulator of interferon genes (cGAS-STING), and that inhibition of the cGAS-STING pathway could be beneficial. It is worth noting that DNase I markedly suppressed the activation of cGAS-STING, which was reversed by the cGAS product cyclic guanosine monophosphate-adenosine monophosphate (cGMP-AMP, cGAMP). Furthermore, the neuroprotective effect of DNase I in SBI was also abolished by cGAMP. NETs may participate in the pathophysiological regulation of SBI by acting through the cGAS-STING pathway. We also found that high-dose vitamin C administration could effectively inhibit the formation of NETs post-SBI. Thus, targeting NETs may provide a novel therapeutic strategy for SBI treatment, and high-dose vitamin C intervention may be a promising translational therapy with an excellent safety profile and low cost.


Subject(s)
Brain Injuries , Extracellular Traps , Animals , Rats , Brain , Brain Injuries/drug therapy , Ascorbic Acid , Deoxyribonuclease I/pharmacology
6.
J Neurosci Res ; 102(4): e25329, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38597144

ABSTRACT

There is a need for new treatments to reduce brain injuries derived from neonatal hypoxia/ischemia. The only viable option used in the clinic today in infants born at term is therapeutic hypothermia, which has a limited efficacy. Treatments with exogenous RNase have shown great promise in a range of different adult animal models including stroke, ischemia/reperfusion injury, or experimental heart transplantation, often by conferring vascular protective and anti-inflammatory effects. However, any neuroprotective function of RNase treatment in the neonate remains unknown. Using a well-established model of neonatal hypoxic/ischemic brain injury, we evaluated the influence of RNase treatment on RNase activity, gray and white matter tissue loss, blood-brain barrier function, as well as levels and expression of inflammatory cytokines in the brain up to 6 h after the injury using multiplex immunoassay and RT-PCR. Intraperitoneal treatment with RNase increased RNase activity in both plasma and cerebropinal fluids. The RNase treatment resulted in a reduction of brain tissue loss but did not affect the blood-brain barrier function and had only a minor modulatory effect on the inflammatory response. It is concluded that RNase treatment may be promising as a neuroprotective regimen, whereas the mechanistic effects of this treatment appear to be different in the neonate compared to the adult and need further investigation.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Infant, Newborn , Infant , Humans , Animals, Newborn , Ribonucleases/metabolism , Ribonucleases/pharmacology , Brain Injuries/drug therapy , Brain/metabolism , Ischemia/drug therapy , Neuroprotective Agents/pharmacology , Disease Models, Animal
7.
Neuropharmacology ; 251: 109896, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38490299

ABSTRACT

Secondary brain injury after intracerebral hemorrhage (ICH) is the main cause of poor prognosis in ICH patients, but the underlying mechanisms remain less known. The involvement of Piezo1 in brain injury after ICH was studied in a mouse model of ICH. ICH was established by injecting autologous arterial blood into the basal ganglia in mice. After vehicle, Piezo1 blocker, GsMTx4, Piezo1 activator, Yoda-1, or together with mannitol (tail vein injection) was injected into the left lateral ventricle of mouse brain, Piezo1 level and the roles of Piezo1 in neuronal injury, brain edema, and neurological dysfunctions after ICH were determined by the various indicated methods. Piezo1 protein level in neurons was significantly upregulated 24 h after ICH in vivo (human and mice). Piezo1 protein level was also dramatically upregulated in HT22 cells (a murine neuron cell line) cultured in vitro 24 h after hemin treatment as an in vitro ICH model. GsMTx4 treatment or together with mannitol significantly downregulated Piezo1 and AQP4 levels, markedly increased Bcl2 level, maintained more neurons alive, considerably restored brain blood flow, remarkably relieved brain edema, substantially decreased serum IL-6 level, and almost fully reversed the neurological dysfunctions at ICH 24 h group mice. In contrast, Yoda-1 treatment achieved the opposite effects. In conclusion, Piezo1 plays a crucial role in the pathogenesis of brain injury after ICH and may be a target for clinical treatment of ICH.


Subject(s)
Brain Edema , Brain Injuries , Pyrazines , Thiadiazoles , Humans , Mice , Animals , Cerebral Hemorrhage/complications , Brain Injuries/drug therapy , Ion Channels , Brain Edema/metabolism , Mannitol/therapeutic use
8.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38474253

ABSTRACT

The brain's unique characteristics make it exceptionally susceptible to oxidative stress, which arises from an imbalance between reactive oxygen species (ROS) production, reactive nitrogen species (RNS) production, and antioxidant defense mechanisms. This review explores the factors contributing to the brain's vascular tone's vulnerability in the presence of oxidative damage, which can be of clinical interest in critically ill patients or those presenting acute brain injuries. The brain's high metabolic rate and inefficient electron transport chain in mitochondria lead to significant ROS generation. Moreover, non-replicating neuronal cells and low repair capacity increase susceptibility to oxidative insult. ROS can influence cerebral vascular tone and permeability, potentially impacting cerebral autoregulation. Different ROS species, including superoxide and hydrogen peroxide, exhibit vasodilatory or vasoconstrictive effects on cerebral blood vessels. RNS, particularly NO and peroxynitrite, also exert vasoactive effects. This review further investigates the neuroprotective effects of antioxidants, including superoxide dismutase (SOD), vitamin C, vitamin E, and the glutathione redox system. Various studies suggest that these antioxidants could be used as adjunct therapies to protect the cerebral vascular tone under conditions of high oxidative stress. Nevertheless, more extensive research is required to comprehensively grasp the relationship between oxidative stress and cerebrovascular tone, and explore the potential benefits of antioxidants as adjunctive therapies in critical illnesses and acute brain injuries.


Subject(s)
Brain Injuries , Oxygen , Humans , Reactive Oxygen Species/metabolism , Oxygen/pharmacology , Nitrogen/pharmacology , Oxidative Stress , Antioxidants/pharmacology , Reactive Nitrogen Species/metabolism , Niacinamide/pharmacology , Brain Injuries/drug therapy
9.
Pharmacol Res Perspect ; 12(2): e1181, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38429943

ABSTRACT

Our laboratory has shown that calpain-2 activation in the brain following acute injury is directly related to neuronal damage and the long-term functional consequences of the injury, while calpain-1 activation is generally neuroprotective and calpain-1 deletion exacerbates neuronal injury. We have also shown that a relatively selective calpain-2 inhibitor, referred to as C2I, enhanced long-term potentiation and learning and memory, and provided neuroprotection in the controlled cortical impact (CCI) model of traumatic brain injury (TBI) in mice. Using molecular dynamic simulation and Site Identification by Ligand Competitive Saturation (SILCS) software, we generated about 130 analogs of C2I and tested them in a number of in vitro and in vivo assays. These led to the identification of two interesting compounds, NA-112 and NA-184. Further analyses indicated that NA-184, (S)-2-(3-benzylureido)-N-((R,S)-1-((3-chloro-2-methoxybenzyl)amino)-1,2-dioxopentan-3-yl)-4-methylpentanamide, selectively and dose-dependent inhibited calpain-2 activity without evident inhibition of calpain-1 at the tested concentrations in mouse brain tissues and human cell lines. Like NA-112, NA-184 inhibited TBI-induced calpain-2 activation and cell death in mice and rats, both male and females. Pharmacokinetic and pharmacodynamic analyses indicated that NA-184 exhibited properties, including stability in plasma and liver and blood-brain barrier permeability, that make it a good clinical candidate for the treatment of TBI.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Neuroprotective Agents , Animals , Humans , Male , Mice , Rats , Brain/metabolism , Brain Injuries/drug therapy , Brain Injuries, Traumatic/drug therapy , Calpain/antagonists & inhibitors , Neuroprotection , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology
10.
Phytomedicine ; 128: 155529, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38503156

ABSTRACT

BACKGROUND/PURPOSE: Rhodiola crenulata (Hook. f. et Thoms.) H. Ohba (R. crenulate), a famous and characteristic Tibetan medicine, has been demonstrated to exert an outstanding brain protection role in the treatment of high-altitude hypoxia disease. However, the metabolic effects of R. crenulate on high-altitude hypoxic brain injury (HHBI) are still incompletely understood. Herein, the anti-hypoxic effect and associated mechanisms of R. crenulate were explored through both in vivo and in vitro experiments. STUDY DESIGN/METHODS: The mice model of HHBI was established using an animal hypobaric and hypoxic chamber. R. crenulate extract (RCE, 0.5, 1.0 and 2.0 g/kg) and salidroside (Sal, 25, 50 and 100 mg/kg) was given by gavage for 7 days. Pathological changes and neuronal apoptosis of mice hippocampus and cortex were evaluated using H&E and TUNEL staining, respectively. The effects of RCE and Sal on the permeability of blood brain barrier (BBB) were detected by Evans blue staining and NIR-II fluorescence imaging. Meanwhile, the ultrastructural BBB and cerebrovascular damages were observed using a transmission electron microscope (TEM). The levels of tight junction proteins Claudin-1, ZO-1 and occludin were detected by immunofluorescence. Additionally, the metabolites in mice serum and brain were determined using UHPLC-MS and MALDI-MSI analysis. The cell viability of Sal on hypoxic HT22 cells induced by CoCl2 was investigated by cell counting kit-8. The contents of LDH, MDA, SOD, GSH-PX and SDH were detected by using commercial biochemical kits. Meanwhile, intracellular ROS, Ca2+ and mitochondrial membrane potential were determined by corresponding specific labeled probes. The intracellular metabolites of HT22 cells were performed by the targeted metabolomics analysis of the Q300 kit. The cell apoptosis and necrosis were examined by YO-PRO-1/PI, Annexin V/PI and TUNEL staining. In addition, mitochondrial morphology was tested by Mito-tracker red with confocal microscopy and TEM. Real-time ATP production, oxygen consumption rate, and proton efflux rate were measured using a Seahorse analyzer. Subsequently, MCU, OPA1, p-Drp1ser616, p-AMPKα, p-AMPKß and Sirt1 were determined by immunofluorescent and western blot analyses. RESULTS: The results demonstrated that R. crenulate and Sal exert anti-hypoxic brain protection from inhibiting neuronal apoptosis, maintaining BBB integrity, increasing tight junction protein Claudin-1, ZO-1 and occludin and improving mitochondrial morphology and function. Mechanistically, R. crenulate and Sal alleviated HHBI by enhancing the tricarboxylic acid cycle to meet the demand of energy of brain. Additionally, experiments in vitro confirmed that Sal could ameliorate the apoptosis of HT22 cells, improve mitochondrial morphology and energy metabolism by enhancing mitochondrial respiration and glycolysis. Meanwhile, Sal-mediated MCU inhibited the activation of Drp1 and enhanced the expression of OPA1 to maintain mitochondrial homeostasis, as well as activation of AMPK and Sirt1 to enhance ATP production. CONCLUSION: Collectively, the findings suggested that RCE and Sal may afford a protective intervention in HHBI through maintaining BBB integrity and improving energy metabolism via balancing MCU-mediated mitochondrial homeostasis by activating the AMPK/Sirt1 signaling pathway.


Subject(s)
Blood-Brain Barrier , Energy Metabolism , Plant Extracts , Rhodiola , Animals , Rhodiola/chemistry , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Mice , Plant Extracts/pharmacology , Energy Metabolism/drug effects , Male , Apoptosis/drug effects , Glucosides/pharmacology , Disease Models, Animal , Phenols/pharmacology , Brain Injuries/drug therapy , Brain Injuries/metabolism , Cell Line , Mitochondria/drug effects , Mitochondria/metabolism , Altitude Sickness/drug therapy , Altitude Sickness/metabolism , Hypoxia/drug therapy
11.
J Ethnopharmacol ; 328: 118114, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38552993

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Alcohol misuse persists as a prevalent societal concern and precipitates diverse deleterious consequences, entailing significant associated health hazards including acute alcohol intoxication (AAI). Binge drinking, a commonplace pattern of alcohol consumption, may incite neurodegeneration and neuronal dysfunction. Clinicians tasked with managing AAI confront a dearth of pharmaceutical intervention alternatives. In contrast, natural products have garnered interest due to their compatibility with the human body and fewer side effects. Lingjiao Gouteng decoction (LGD), a classical traditional Chinese medicine decoction, represents a frequently employed prescription in cases of encephalopathy, although its efficacy in addressing acute alcoholism and alcohol-induced brain injury remains inadequately investigated. AIM OF THE STUDY: To investigate the conceivable therapeutic benefits of LGD in AAI and alcohol-induced brain injury, while delving into the underlying fundamental mechanisms involved. MATERIALS AND METHODS: We established an AAI mouse model through alcohol gavage, and LGD was administered to the mice twice at the 2 h preceding and 30 min subsequent to alcohol exposure. The study encompassed the utilization of the loss of righting reflex assay, histopathological analysis, enzyme-linked immunosorbent assays, and cerebral tissue biochemical assays to investigate the impact of LGD on AAI and alcohol-induced brain injury. These assessments included a comprehensive evaluation of various biomarkers associated with the inflammatory response and oxidative stress. Finally, RT-qPCR, Western blot, and immunofluorescence staining were carried out to explore the underlying mechanisms through which LGD exerts its therapeutic influence, potentially through the regulation of the RhoA/ROCK2/NF-κB signaling pathway. RESULTS: Our investigation underscores the therapeutic efficacy of LGD in ameliorating AAI, as evidenced by discernible alterations in the loss of righting reflex assay, pathological analysis, and assessment of inflammatory and oxidative stress biomarkers. Furthermore, the results of RT-qPCR, Western blot, and immunofluorescence staining manifest a noteworthy regulatory effect of LGD on the RhoA/ROCK2/NF-κB signaling pathway. CONCLUSIONS: The present study confirmed the therapeutic potential of LGD in AAI and alcohol-induced brain injury, and the protective effects of LGD against alcohol-induced brain injury may be intricately linked to the RhoA/ROCK2/NF-κB signaling pathway.


Subject(s)
Alcoholic Intoxication , Alcoholism , Brain Injuries , Mice , Humans , Animals , NF-kappa B/metabolism , Alcoholic Intoxication/drug therapy , Signal Transduction , Ethanol/pharmacology , Brain Injuries/drug therapy , Biomarkers , rho-Associated Kinases/metabolism
12.
Neurochem Int ; 175: 105676, 2024 May.
Article in English | MEDLINE | ID: mdl-38336256

ABSTRACT

BACKGROUND: Microglia-mediated neuroinflammation is the major contributor to the secondary brain injury of ischemic stroke. NLRP3 is one of the major components of ischemia-induced microglial activation. Echinatin, a chalcone found in licorice, was reported to have the activity of anti-inflammation and antioxidant. However, the relative study of echinatin in microglia or ischemic stroke is still unclear. METHODS: We intravenously injected echinatin or vehicle into adult ischemic male C57/BL6J mice induced by 60-min transient middle cerebral artery occlusion (tMCAO). The intraperitoneal injection was performed 4.5 h after reperfusion and then daily for 2 more days. Infarct size, blood brain barrier (BBB) leakage, neurobehavioral tests, and microglial-mediated inflammatory reaction were examined to assess the outcomes of echinatin treatment. LPS and LPS/ATP stimulation on primary microglia were used to explore the underlying anti-inflammatory mechanism of echinatin. RESULTS: Echinatin treatment efficiently decreased the infarct size, alleviated blood brain barrier (BBB) damage, suppressed microglial activation, reduced the production of inflammatory factors (e.g., IL-1ß, IL-6, IL-18, TNF-α, iNOS, COX2), and relieved post-stroke neurological defects in tMCAO mice. Mechanistically, we found that echinatin could suppress the NLRP3 assembly and reduce the production of inflammatory mediators independently of NF-κB and monoamine oxidase (MAO). CONCLUSION: Based on our study, we have identified echinatin as a promising therapeutic strategy for the treatment of ischemic stroke.


Subject(s)
Brain Injuries , Brain Ischemia , Chalcones , Ischemic Stroke , Reperfusion Injury , Mice , Male , Animals , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Lipopolysaccharides , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/complications , Infarction/complications , Infarction/drug therapy , Anti-Inflammatory Agents/therapeutic use , Brain Injuries/drug therapy , Ischemic Stroke/drug therapy , Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , Brain Ischemia/complications , Microglia , Reperfusion Injury/drug therapy
13.
Brain Inj ; 38(5): 337-340, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38308526

ABSTRACT

BACKGROUND: In rare cases, zolpidem administration has been found to paradoxically improve cognition in patients with brain injury in disorders of consciousness. CASE PRESENTATION: Two minimally conscious plus (MCS+) patients at baseline, a 24-year-old woman 8 weeks post-traumatic brain injury (TBI) and 23-year-old man 6 weeks post-TBI, demonstrated behavioral improvements after off-label, single-dose administration of 10 mg of zolpidem. DISCUSSION/CONCLUSION: The patients demonstrated improved cognition on Coma Recovery Scale-Revised assessment after ingesting zolpidem. In particular, speech was substantially restored as one patient recovered functional communication and both demonstrated intelligible verbalizations for the first-time post-injuries following zolpidem. Overall, evidence is limited regarding the underlying mechanisms of various cognitive improvements in zolpidem response although studies incorporating neuroimaging are promising. The outcomes and similarities between these cases contribute to the current literature and highlight the need for rigorous studies in the future to guide zolpidem trials in patient care for those with DOC.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Chronic Traumatic Encephalopathy , Male , Female , Humans , Young Adult , Adult , Zolpidem , Speech , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries/complications , Brain Injuries/drug therapy , Persistent Vegetative State/drug therapy , Persistent Vegetative State/etiology , Consciousness Disorders/drug therapy , Consciousness Disorders/etiology , Chronic Traumatic Encephalopathy/complications , Recovery of Function/physiology
14.
J Clin Monit Comput ; 38(1): 25-30, 2024 02.
Article in English | MEDLINE | ID: mdl-38310591

ABSTRACT

Brain injury patients require precise blood pressure (BP) management to maintain cerebral perfusion pressure (CPP) and avoid intracranial hypertension. Nurses have many tasks and norepinephrine titration has been shown to be suboptimal. This can lead to limited BP control in patients that are in critical need of cerebral perfusion optimization. We have designed a closed-loop vasopressor (CLV) system capable of maintaining mean arterial pressure (MAP) in a narrow range and we aimed to assess its performance when treating severe brain injury patients. Within the first 48 h of intensive care unit (ICU) admission, 18 patients with a severe brain injury underwent either CLV or manual norepinephrine titration. In both groups, the objective was to maintain MAP in target (within ± 5 mmHg of a predefined target MAP) to achieve optimal CPP. Fluid administration was standardized in the two groups. The primary objective was the percentage of time patients were in target. Secondary outcomes included time spent over and under target. Over the four-hour study period, the mean percentage of time with MAP in target was greater in the CLV group than in the control group (95.8 ± 2.2% vs. 42.5 ± 27.0%, p < 0.001). Severe undershooting, defined as MAP < 10 mmHg of target value was lower in the CLV group (0.2 ± 0.3% vs. 7.4 ± 14.2%, p < 0.001) as was severe overshooting defined as MAP > 10 mmHg of target (0.0 ± 0.0% vs. 22.0 ± 29.0%, p < 0.001). The CLV system can maintain MAP in target better than nurses caring for severe brain injury patients.


Subject(s)
Brain Injuries , Norepinephrine , Humans , Arterial Pressure , Vasoconstrictor Agents/therapeutic use , Brain Injuries/drug therapy , Intensive Care Units , Intracranial Pressure
15.
Cell Mol Biol (Noisy-le-grand) ; 70(1): 19-27, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38372118

ABSTRACT

Sepsis is regarded as an inflammatory syndrome that consists of complex biochemical and pathophysiological dysregulation, brought on by endogenous factors in response to systemic infection. Sepsis can cause short- and long-term cerebral injury. Cerium oxide nanoparticles (CeO2 NPs) have been reported to possess both anti-inflammatory and antioxidative properties. The current study investigated the potential role of cerium oxide nanoparticles in the management of sepsis-induced brain injury. To achieve this target, forty male albino rats were divided into 4 groups, ten rats each. Group (i) set as a shame group. Group (ii) set as shame group administrated CeO2 NPs. Group (iii) septic group treated with saline and Group (iv) septic group treated with CeO2 NPs. The sepsis model in rats was induced by cecal ligation and puncture (CLP). Results showed CeO2 NPs administration resulted in significant improvement in the survival rate of rats, suppression in serum sepsis biomarkers (CRP, ESM-1, PCT and D- dimer), amelioration of brain inflammatory mediators (TNF-α- IL-6, NF-kB and LTB4) as well as apoptotic markers (Cas-3 and BAX). Furthermore, immunomodulation of miRNAs expression (155,124 and 146a). These findings demonstrate a promising pivotal role of CeO2 NPs treatment in alleviating the deleterious effects induced by sepsis in the brain.


Subject(s)
Brain Injuries , Cerium , MicroRNAs , Nanoparticles , Sepsis , Rats , Male , Animals , NF-kappa B/metabolism , MicroRNAs/genetics , Nanoparticles/chemistry , Brain Injuries/drug therapy , Sepsis/complications , Sepsis/drug therapy
16.
Eur Rev Med Pharmacol Sci ; 28(3): 1194-1201, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38375724

ABSTRACT

OBJECTIVE: This study aimed to investigate the association between serum galanin (GAL) and neuron-specific enolase (NSE) levels in children with convulsive status epilepticus (CSE) and their relationship with abnormal electroencephalogram (EEG) patterns. Additionally, the study assessed the effectiveness of a combination therapy involving midazolam, diazepam, and phenobarbital in treating CSE. PATIENTS AND METHODS: The research involved 100 children diagnosed with CSE and included a control group of 50 healthy children. Serum GAL and NSE levels were measured, and EEGs were analyzed for abnormalities in the CSE group. Comparisons were made between the healthy control group and the CSE group, particularly within the first 24 hours after persistent seizures. The severity of EEG abnormalities was correlated with GAL and NSE levels. The treatment consisted of an observation group that received the triple therapy of midazolam, diazepam, and phenobarbital, while a control group received diazepam and phenobarbital. Clinical efficacy, symptom improvement, Status Epilepticus Severity Score (STESS), and adverse reactions were evaluated. RESULTS: The results indicated elevated levels of GAL and NSE in the CSE group, with higher levels noted within 24 hours after persistent seizures. Furthermore, a positive correlation was observed between the severity of EEG abnormalities and GAL and NSE levels. The group receiving the triple therapy demonstrated superior efficacy, faster resolution of seizures and fever, reduced STESS scores, and fewer adverse reactions than the control group. In conclusion, this study highlights the positive correlation between serum GAL and NSE levels and the severity of EEG abnormalities in pediatric CSE. The triple therapy approach is effective in treating CSE, leading to improved clinical symptoms, reduced brain damage, and enhanced safety. CONCLUSIONS: The study concludes that serum GAL and NSE levels in children with convulsive status epilepticus are positively correlated with the degree of EEG abnormalities. The combination therapy involving midazolam, diazepam, and phenobarbital is effective in treating children with convulsive status epilepticus, significantly improving clinical symptoms, reducing brain damage, and ensuring safety.


Subject(s)
Brain Injuries , Status Epilepticus , Child , Humans , Midazolam/therapeutic use , Galanin , Status Epilepticus/diagnosis , Status Epilepticus/drug therapy , Seizures/drug therapy , Diazepam/therapeutic use , Phenobarbital/therapeutic use , Electroencephalography , Brain Injuries/drug therapy , Phosphopyruvate Hydratase , Anticonvulsants/therapeutic use
17.
Mol Biol Rep ; 51(1): 366, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409545

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) is one of the most prevalent brain injuries in humans which has poor prognosis and high mortality rates. Due to several medical or surgical treatment methods, a gold standard method doesn't exist for SAH treatment. Piceatannol (PCN), a natural analog of resveratrol, was reported to reduce inflammation and apoptosis promising a wide range of therapeutic alternatives. In this study, we aimed to investigate the effects of PCN in an experimental SAH model. The alleviating effects of PCN in the hippocampus in an experimental SAH model were investigated for the first time. METHODS AND RESULTS: In this study, 27 Wistar Albino male rats (200-300 g; 7-8 week) were used. Animals were divided into three groups; SHAM, SAH, and SAH + PCN. SAH model was created with 120 µl of autologous arterial tail blood to prechiasmatic cisterna. 30 mg/kg PCN was administered intraperitoneally at 1st h after SAH. Neurological evaluation was performed with Garcia's score. RT-PCR was performed for gene expression levels in the hippocampus. Pyknosis, edema, and apoptosis were evaluated by H&E and TUNEL staining. Our results indicated that PCN administration reduced apoptosis (P < 0.01), cellular edema, and pyknosis (P < 0.05) in the hippocampus after SAH. Moreover, PCN treatment significantly decreased the expression levels of TNF-α (P < 0.01), IL-6 (P < 0.05), NF-κB (P < 0.05), and Bax (P < 0.05) in the hippocampus. CONCLUSIONS: Our results demonstrated that PCN might be a potential therapeutic adjuvant agent for the treatment of early brain injury (EBI) following SAH. Further studies are required to clarify the underlying mechanisms and treatment options of SAH.


Subject(s)
Brain Injuries , Neuroprotective Agents , Stilbenes , Subarachnoid Hemorrhage , Humans , Rats , Animals , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Rats, Wistar , Brain Injuries/drug therapy , Apoptosis , Edema/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
18.
Biomaterials ; 306: 122495, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38309053

ABSTRACT

In managing severe traumatic brain injury (TBI), emergency surgery involving the removal of damaged brain tissue and intracerebral hemorrhage is a priority. Secondary brain injury caused by oxidative stress and energy metabolic disorders, triggered by both primary mechanical brain damage and surgical insult, is also a determining factor in the prognosis of TBI. Unfortunately, the effectiveness of traditional postoperative intravenous neuroprotective agents therapy is often limited by the lack of targeting, timeliness, and side effects when neuroprotective agents systemically delivered. Here, we have developed injectable, intelligent, self-assembling hydrogels (P-RT/2DG) that can achieve precise treatment through intraoperative application to the target area. P-RT/2DG hydrogels were prepared by integrating a reactive oxygen species (ROS)-responsive thioketal linker (RT) into polyethylene glycol. By scavenging ROS and releasing 2-deoxyglucose (2DG) during degradation, these hydrogels functioned both in antioxidation and energy metabolism to inhibit the vicious cycle of post-TBI ROS-lactate which provoked secondary injury. In vitro and in vivo tests confirmed the absence of systemic side effects and the neuroprotective function of P-RT/2DG hydrogels in reducing edema, nerve cell apoptosis, neuroinflammation, and maintaining the blood-brain barrier. Our study thus provides a potential treatment strategy with novel hydrogels in TBI.


Subject(s)
Brain Injuries , Neuroprotective Agents , Humans , Reactive Oxygen Species/metabolism , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Hydrogels/pharmacology , Brain/metabolism , Brain Injuries/drug therapy , Energy Metabolism
19.
Molecules ; 29(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38398528

ABSTRACT

Kaempferol, a flavonoid present in many food products, has chemical and cellular antioxidant properties that are beneficial for protection against the oxidative stress caused by reactive oxygen and nitrogen species. Kaempferol administration to model experimental animals can provide extensive protection against brain damage of the striatum and proximal cortical areas induced by transient brain cerebral ischemic stroke and by 3-nitropropionic acid. This article is an updated review of the molecular and cellular mechanisms of protection by kaempferol administration against brain damage induced by these insults, integrated with an overview of the contributions of the work performed in our laboratories during the past years. Kaempferol administration at doses that prevent neurological dysfunctions inhibit the critical molecular events that underlie the initial and delayed brain damage induced by ischemic stroke and by 3-nitropropionic acid. It is highlighted that the protection afforded by kaempferol against the initial mitochondrial dysfunction can largely account for its protection against the reported delayed spreading of brain damage, which can develop from many hours to several days. This allows us to conclude that kaempferol administration can be beneficial not only in preventive treatments, but also in post-insult therapeutic treatments.


Subject(s)
Brain Injuries , Ischemic Stroke , Neuroprotective Agents , Nitro Compounds , Propionates , Stroke , Animals , Kaempferols/pharmacology , Brain , Oxidative Stress , Stroke/drug therapy , Ischemia/drug therapy , Brain Injuries/drug therapy , Reperfusion , Ischemic Stroke/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
20.
Stroke ; 55(3): 725-734, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38406851

ABSTRACT

BACKGROUND: Remote secondary neurodegeneration is associated with poststroke cognitive impairment (PSCI). Dl-3-n-butylphthalide (NBP) improves PSCI clinically. However, whether it ameliorates PSCI by alleviating secondary neurodegeneration remains uncertain. Nonhuman primates provide more relevant models than rodents for human stroke and PSCI. This study investigated the effects of NBP on PSCI and secondary neurodegeneration in cynomolgus monkeys after permanent left middle cerebral artery occlusion (MCAO). METHODS: Thirteen adult male cynomolgus monkeys were randomly assigned to sham (n=4), MCAO+placebo (n=5), and MCAO+NBP groups (n=4). The MCAO+placebo and MCAO+NBP groups received saline and NBP injections intravenously, respectively, starting at 6-hour postsurgery for 2 weeks, followed by soybean oil and NBP orally, respectively, for 10 weeks after MCAO. Infarct size was assessed at week 4 by magnetic resonance imaging. Working memory and executive function were evaluated dynamically using the delayed response task and object retrieval detour task, respectively. Neuron loss, glia proliferation, and neuroinflammation in the ipsilateral dorsal lateral prefrontal cortex, thalamus, and hippocampus were analyzed by immunostaining 12 weeks after MCAO. RESULTS: Infarcts were located in the left middle cerebral artery region, apart from the ipsilateral dorsal lateral prefrontal cortex, thalamus, or hippocampus, with no significant difference between the MCAO+placebo and MCAO+NBP group. Higher success in delayed response task was achieved at weeks 4, 8, and 12 after NBP compared with placebo treatments (P<0.05), but not in the object retrieval detour task (all P>0.05). More neurons and less microglia, astrocytes, CD68-positive microglia, tumor necrosis factor-α, and inducible NO synthase were observed in the ipsilateral dorsal lateral prefrontal cortex and thalamus after 12 weeks of NBP treatment (P<0.05), but not in the hippocampus (P>0.05). CONCLUSIONS: Our findings indicate that NBP improves working memory by alleviating remote secondary neurodegeneration and neuroinflammation in the ipsilateral dorsal lateral prefrontal cortex and thalamus after MCAO in cynomolgus monkeys.


Subject(s)
Benzofurans , Brain Injuries , Brain Neoplasms , Neuroprotective Agents , Stroke , Humans , Animals , Male , Macaca fascicularis , Memory, Short-Term , Neuroinflammatory Diseases , Stroke/complications , Stroke/diagnostic imaging , Stroke/drug therapy , Brain Injuries/drug therapy , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Hippocampus/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...