Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.686
Filtrer
1.
J Biochem Mol Toxicol ; 38(7): e23760, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953502

RÉSUMÉ

Cyclophosphamide (CP) is an antineoplastic drug widely used in chemotherapy. Curcumin (CUR) and piperine (PP) show a protective effect on neurodegenerative and neurological diseases. This research was designed to measure several biochemical parameters in the brain tissue of CP-applied rats to investigate the impact of combined CUR-PP administration. The study evaluated six groups of eight rats: Group 1 was the control; Groups 2 and 3 were administered 200 or 300 mg/kg CUR-PP via oral gavage; Group 4 received only 200 mg/kg CP on day 1; Groups 5 and 6 received CP + CUR-PP for 7 days. Data from all parameters indicated that CP caused brain damage. Phosphorylated TAU (pTAU), amyloid-beta peptide 1-42 (Aß1-42), glutamate (GLU), and gamma amino butyric acid (GABA) parameters were the same in Groups 4, 5, and 6. On the other hand, 8-hydroxy-2-deoxyguanosine (8-OHdG), nitric oxide (NO), interleukin-6 (IL-6), nuclear factor kappa beta (NF-kß), malondialdehyde (MDA), and tumor necrosis factor-alpha (TNF-α) levels in the CP + CUR-PP groups were lower than those in the CP group (p < 0.05). However, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and reduced glutathione (GSH) parameters were higher in the CP + CUR-PP groups compared to the CP group (p < 0.05). It is thought that the similarity of Groups 5 and 6 with Group 4 in Aß1-42, pTAU, GLU, and GABA parameters hinder the determination of treatment protection however, they might have a therapeutic effect if the applied dose or study duration were changed. This study attempted to evaluate the effects of a CUR-PP combination on CP-induced brain damage in rats by measuring biochemical parameters and performing histopathological examinations. Based on the findings, this CUR-PP combination could be considered an alternative medicine option in cases with conditions similar to those evaluated in this study.


Sujet(s)
Alcaloïdes , Benzodioxoles , Lésions encéphaliques , Curcumine , Cyclophosphamide , Pipéridines , Amides gras polyinsaturés N-alkylés , Animaux , Amides gras polyinsaturés N-alkylés/pharmacologie , Benzodioxoles/pharmacologie , Curcumine/pharmacologie , Pipéridines/pharmacologie , Alcaloïdes/pharmacologie , Rats , Cyclophosphamide/toxicité , Cyclophosphamide/effets indésirables , Mâle , Lésions encéphaliques/induit chimiquement , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , Lésions encéphaliques/anatomopathologie , Lésions encéphaliques/prévention et contrôle , Rat Wistar , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie
2.
Int J Biol Macromol ; 273(Pt 1): 132887, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38851621

RÉSUMÉ

Acute alcohol intoxication is a harmful clinical condition characterized by behavioral and neurological symptoms, for which few effective therapies are available at present. Dysfunction of microglial BV-2 cells has been reported to be associated with acute alcohol-induced brain injuries. In the present study, the protective effects of Eucommia ulmoides Oliv. leaves polysaccharides (EULP) on acute alcoholic brain injury and microglial dysfunction were investigated. 14-day pretreatment of EULP significantly attenuated neurobehavioral deficit and neurotransmitter damage in the brain tissue of mice caused by acute alcohol exposure. Additionally, EULP regulated the metabolic disorder of brain tissue. Consistently, it was shown that EULP pretreatment significantly improved alcohol-induced phagocytosis decrease, oxidative stress and inflammation in BV-2 cells. Therefore, EULP may be proposed and employed as a potential therapeutic agent for alcohol-induced brain damage.


Sujet(s)
Eucommiaceae , Microglie , Stress oxydatif , Feuilles de plante , Polyosides , Animaux , Polyosides/pharmacologie , Polyosides/composition chimique , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Souris , Eucommiaceae/composition chimique , Feuilles de plante/composition chimique , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Éthanol , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/induit chimiquement , Lésions encéphaliques/métabolisme , Lésions encéphaliques/étiologie , Lésions encéphaliques/anatomopathologie , Lignée cellulaire , Phagocytose/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique
3.
EMBO Mol Med ; 16(6): 1228-1253, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38789599

RÉSUMÉ

In the injured brain, new neurons produced from endogenous neural stem cells form chains and migrate to injured areas and contribute to the regeneration of lost neurons. However, this endogenous regenerative capacity of the brain has not yet been leveraged for the treatment of brain injury. Here, we show that in healthy brain chains of migrating new neurons maintain unexpectedly large non-adherent areas between neighboring cells, allowing for efficient migration. In instances of brain injury, neuraminidase reduces polysialic acid levels, which negatively regulates adhesion, leading to increased cell-cell adhesion and reduced migration efficiency. The administration of zanamivir, a neuraminidase inhibitor used for influenza treatment, promotes neuronal migration toward damaged regions, fosters neuronal regeneration, and facilitates functional recovery. Together, these findings shed light on a new mechanism governing efficient neuronal migration in the adult brain under physiological conditions, pinpoint the disruption of this mechanism during brain injury, and propose a promising therapeutic avenue for brain injury through drug repositioning.


Sujet(s)
Encéphale , Mouvement cellulaire , Sialidase , Neurones , Sialidase/métabolisme , Sialidase/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , Animaux , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Souris , Zanamivir/pharmacologie , Antienzymes/pharmacologie , Acides sialiques/métabolisme , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Adhérence cellulaire/effets des médicaments et des substances chimiques , Humains , Mâle
4.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 220-225, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38814212

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Facteur neurotrophique dérivé du cerveau , Cognition , Protéine de liaison à l'élément de réponse à l'AMP cyclique , AMP cyclique , Quinuclidines , Rat Sprague-Dawley , Transduction du signal , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Quinuclidines/pharmacologie , Quinuclidines/usage thérapeutique , Cognition/effets des médicaments et des substances chimiques , Mâle , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , AMP cyclique/métabolisme , Rats , Facteur de croissance nerveuse/métabolisme , Facteur de croissance nerveuse/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Modèles animaux de maladie humaine
5.
Am J Chin Med ; 52(3): 799-819, 2024.
Article de Anglais | MEDLINE | ID: mdl-38752843

RÉSUMÉ

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.


Sujet(s)
Alcaloïdes indoliques , Facteur de transcription NF-kappa B , Quinazolines , Rat Sprague-Dawley , Sirtuines , Hémorragie meningée , Animaux , Hémorragie meningée/traitement médicamenteux , Hémorragie meningée/complications , Sirtuines/métabolisme , Sirtuines/génétique , Alcaloïdes indoliques/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Mâle , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Modèles animaux de maladie humaine , Lésions encéphaliques/étiologie , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , Rats , Inflammation/traitement médicamenteux , Inflammation/étiologie , Phytothérapie , Transduction du signal/effets des médicaments et des substances chimiques , Expression des gènes/effets des médicaments et des substances chimiques , Quinazolinones
6.
Front Immunol ; 15: 1386780, 2024.
Article de Anglais | MEDLINE | ID: mdl-38756773

RÉSUMÉ

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.


Sujet(s)
Hémorragie cérébrale , MAP Kinase Kinase Kinases , Facteur-2 apparenté à NF-E2 , Neurones , Stress oxydatif , Pyroptose , Rat Sprague-Dawley , Transduction du signal , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Facteur-2 apparenté à NF-E2/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Hémorragie cérébrale/métabolisme , Hémorragie cérébrale/traitement médicamenteux , Mâle , Rats , Transduction du signal/effets des médicaments et des substances chimiques , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Lésions encéphaliques/étiologie , Lésions encéphaliques/métabolisme , Lésions encéphaliques/traitement médicamenteux , Espèces réactives de l'oxygène/métabolisme , Lactones , Résorcinol , Zéaralénone/administration et posologie
7.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702778

RÉSUMÉ

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.


Sujet(s)
Ferroptose , Souris de lignée C57BL , Maladies neuro-inflammatoires , Hémorragie meningée , Animaux , Hémorragie meningée/métabolisme , Hémorragie meningée/anatomopathologie , Hémorragie meningée/complications , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Souris , Mâle , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/étiologie , Lésions encéphaliques/métabolisme , Lésions encéphaliques/anatomopathologie , Lésions encéphaliques/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie
8.
J Neurosci Res ; 102(4): e25329, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38597144

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Hypoxie-ischémie du cerveau , Neuroprotecteurs , Animaux , Nouveau-né , Nourrisson , Humains , Animaux nouveau-nés , Ribonucléases/métabolisme , Ribonucléases/pharmacologie , Lésions encéphaliques/traitement médicamenteux , Encéphale/métabolisme , Ischémie/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Modèles animaux de maladie humaine
9.
Cell Mol Neurobiol ; 44(1): 36, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637346

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Pièges extracellulaires , Animaux , Rats , Encéphale , Lésions encéphaliques/traitement médicamenteux , Acide ascorbique , Deoxyribonuclease I/pharmacologie
10.
J Ethnopharmacol ; 328: 118114, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38552993

RÉSUMÉ

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.


Sujet(s)
Intoxication alcoolique , Alcoolisme , Lésions encéphaliques , Souris , Humains , Animaux , Facteur de transcription NF-kappa B/métabolisme , Intoxication alcoolique/traitement médicamenteux , Transduction du signal , Éthanol/pharmacologie , Lésions encéphaliques/traitement médicamenteux , Marqueurs biologiques , rho-Associated Kinases/métabolisme
11.
Phytomedicine ; 128: 155529, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38503156

RÉSUMÉ

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.


Sujet(s)
Barrière hémato-encéphalique , Métabolisme énergétique , Extraits de plantes , Rhodiola , Animaux , Rhodiola/composition chimique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Souris , Extraits de plantes/pharmacologie , Métabolisme énergétique/effets des médicaments et des substances chimiques , Mâle , Apoptose/effets des médicaments et des substances chimiques , Glucosides/pharmacologie , Modèles animaux de maladie humaine , Phénols/pharmacologie , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , Lignée cellulaire , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mal de l'altitude/traitement médicamenteux , Mal de l'altitude/métabolisme , Hypoxie/traitement médicamenteux
12.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38474253

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Oxygène , Humains , Espèces réactives de l'oxygène/métabolisme , Oxygène/pharmacologie , Azote/pharmacologie , Stress oxydatif , Antioxydants/pharmacologie , Espèces réactives de l'azote/métabolisme , Nicotinamide/pharmacologie , Lésions encéphaliques/traitement médicamenteux
13.
Pharmacol Res Perspect ; 12(2): e1181, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38429943

RÉSUMÉ

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.


Sujet(s)
Lésions traumatiques de l'encéphale , Lésions encéphaliques , Neuroprotecteurs , Animaux , Humains , Mâle , Souris , Rats , Encéphale/métabolisme , Lésions encéphaliques/traitement médicamenteux , Lésions traumatiques de l'encéphale/traitement médicamenteux , Calpain/antagonistes et inhibiteurs , Neuroprotection , Neuroprotecteurs/composition chimique , Neuroprotecteurs/pharmacologie
14.
Neuropharmacology ; 251: 109896, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38490299

RÉSUMÉ

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.


Sujet(s)
Oedème cérébral , Lésions encéphaliques , Pyrazines , Thiadiazoles , Humains , Souris , Animaux , Hémorragie cérébrale/complications , Lésions encéphaliques/traitement médicamenteux , Canaux ioniques , Oedème cérébral/métabolisme , Mannitol/usage thérapeutique
15.
Molecules ; 29(4)2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-38398528

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Accident vasculaire cérébral ischémique , Neuroprotecteurs , Composés nitrés , Propionates , Accident vasculaire cérébral , Animaux , Kaempférols/pharmacologie , Encéphale , Stress oxydatif , Accident vasculaire cérébral/traitement médicamenteux , Ischémie/traitement médicamenteux , Lésions encéphaliques/traitement médicamenteux , Reperfusion , Accident vasculaire cérébral ischémique/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique
16.
Mol Biol Rep ; 51(1): 366, 2024 Feb 26.
Article de Anglais | MEDLINE | ID: mdl-38409545

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Neuroprotecteurs , Stilbènes , Hémorragie meningée , Humains , Rats , Animaux , Rat Sprague-Dawley , Hémorragie meningée/traitement médicamenteux , Hémorragie meningée/métabolisme , Rat Wistar , Lésions encéphaliques/traitement médicamenteux , Apoptose , Oedème/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique
17.
Biomaterials ; 306: 122495, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38309053

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Neuroprotecteurs , Humains , Espèces réactives de l'oxygène/métabolisme , Neuroprotecteurs/pharmacologie , Oxygène/métabolisme , Hydrogels/pharmacologie , Encéphale/métabolisme , Lésions encéphaliques/traitement médicamenteux , Métabolisme énergétique
18.
Neurochem Int ; 175: 105676, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38336256

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Encéphalopathie ischémique , Chalcones , Accident vasculaire cérébral ischémique , Lésion d'ischémie-reperfusion , Souris , Mâle , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine , Maladies neuro-inflammatoires , Lipopolysaccharides , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/complications , Infarctus/complications , Infarctus/traitement médicamenteux , Anti-inflammatoires/usage thérapeutique , Lésions encéphaliques/traitement médicamenteux , Accident vasculaire cérébral ischémique/traitement médicamenteux , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/prévention et contrôle , Encéphalopathie ischémique/complications , Microglie , Lésion d'ischémie-reperfusion/traitement médicamenteux
19.
Chem Biol Drug Des ; 103(2): e14436, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38395608

RÉSUMÉ

Neuroinflammation contributes to neurological dysfunction in the patients who suffer from subarachnoid hemorrhage (SAH). Isoliquiritigenin (ISL) is a bioactive component extracted from Genus Glycyrrhiza. This work is to investigate whether ISL ameliorates neuroinflammation after SAH. In this study, intravascular perforation of male Sprague-Dawley rats was used to establish a SAH model. ISL was administered by intraperitoneal injection 6 h after SAH in rats. The mortality, SAH grade, neurological score, brain water content, and blood-brain barrier (BBB) permeability were examined at 24 h after the treatment. Expressions of tumor necrosis factor-α, interleukin-6, Iba-1, and MPO were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Besides, the expression levels of NF-κB p65 and NLRP3, ASC, caspase-1, IL-1ß, and IL-18 were analyzed by western blot. The experimental data suggested that ISL treatment could ameliorate neurological impairment, attenuate brain edema, and ameliorate BBB injury after SAH in rats. ISL treatment repressed the expression of proinflammatory cytokines TNF-α and IL-6, and meanwhile inhibited the expression of Iba-1 and MPO. ISL also repressed NF-κB p65 expression as well as the transport from the cytoplasm to the nucleus. In addition, ISL significantly suppressed the expression levels of NLR family pyrin domain containing 3 (NLRP3), ASC, caspase-1, IL-1ß, and IL-18. These findings suggest that ISL inactivates NLRP3 pathway by inhibiting NF-κB p65 translocation, thereby repressing the neuroinflammation after SAH, and it is a potential drug for the treatment of SAH.


Sujet(s)
Lésions encéphaliques , Chalcones , Hémorragie meningée , Humains , Rats , Mâle , Animaux , Facteur de transcription NF-kappa B/métabolisme , Hémorragie meningée/complications , Hémorragie meningée/traitement médicamenteux , Inflammasomes/métabolisme , Rat Sprague-Dawley , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Interleukine-18/usage thérapeutique , Maladies neuro-inflammatoires , Lésions encéphaliques/traitement médicamenteux , Lésions encéphaliques/métabolisme , Lésions encéphaliques/anatomopathologie , Facteur de nécrose tumorale alpha/génétique , Caspases/usage thérapeutique
20.
J Clin Monit Comput ; 38(1): 25-30, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38310591

RÉSUMÉ

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.


Sujet(s)
Lésions encéphaliques , Norépinéphrine , Humains , Pression artérielle , Vasoconstricteurs/usage thérapeutique , Lésions encéphaliques/traitement médicamenteux , Unités de soins intensifs , Pression intracrânienne
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...