Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.936
Filtrer
1.
BMC Complement Med Ther ; 24(1): 293, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090706

RÉSUMÉ

BACKGROUND: Salidroside is the major bioactive and pharmacological active substance in Rhodiola rosea L. It has been reported to have neuroprotective effects on cerebral ischemia/reperfusion (I/R). However, whether salidroside can enhance neural regeneration after cerebral I/R is still unknown. This study investigated the effects of salidroside on the endogenous neural regeneration after cerebral I/R and the related mechanism. METHODS: Focal cerebral I/R was induced in rats by transient middle cerebral artery occlusion/reperfusion (MCAO/R). The rats were intraperitoneally treated salidroside once daily for 7 consecutive days. Neurobehavioral assessments were performed at 3 days and 7 days after the injury. TTC staining was performed to assess cerebral infarct volume. To evaluate the survival of neurons, immunohistochemical staining of Neuronal Nuclei (NeuN) in the ischemic hemisphere were conducted. Also, immunofluorescence double or triple staining of the biomarkers of proliferating neural progenitor cells in Subventricular Zone (SVZ) and striatum of the ischemia hemisphere were performed to investigate the neurogenesis. Furthermore, reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of neurotrophic factors (NTFs) brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). Expression of Notch1 and its target molecular Hes1 were also analyzed by western-blotting and RT-PCR. RESULTS: Salidroside treatment ameliorated I/R induced neurobehavioral impairment, and reduced infarct volume. Salidroside also restored NeuN positive cells loss after I/R injury. Cerebral I/R injury significantly increased the expression of 5-Bromo-2'-Deoxyuridine (BrdU) and doublecotin (DCX), elevated the number of BrdU/Nestin/DCX triple-labeled cells in SVZ, and BrdU/Nestin/glial fibrillary acidic protein (GFAP) triple-labeled cells in striatum. Salidroside treatment further promoted the proliferation of BrdU/DCX labeled neuroblasts and BrdU/Nestin/GFAP labeled reactive astrocytes. Furthermore, salidroside elevated the mRNA expression and protein concentration of BDNF and NGF in ischemia periphery area, as well. Mechanistically, salidroside elevated Notch1/Hes1 mRNA expression in SVZ. The protein levels of them were also increased after salidroside administration. CONCLUSIONS: Salidroside enhances the endogenous neural regeneration after cerebral I/R. The mechanism of the effect may involve the regulation of BDNF/NGF and Notch signaling pathway.


Sujet(s)
Encéphalopathie ischémique , Glucosides , Régénération nerveuse , Phénols , Rat Sprague-Dawley , Lésion d'ischémie-reperfusion , Transduction du signal , Animaux , Glucosides/pharmacologie , Phénols/pharmacologie , Rats , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Lésion d'ischémie-reperfusion/traitement médicamenteux , Encéphalopathie ischémique/traitement médicamenteux , Régénération nerveuse/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Facteurs de croissance nerveuse/métabolisme , Modèles animaux de maladie humaine , Récepteurs Notch/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Neurogenèse/effets des médicaments et des substances chimiques
2.
J Neuroinflammation ; 21(1): 195, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097747

RÉSUMÉ

Chronic cerebral hypoperfusion (CCH), a disease afflicting numerous individuals worldwide, is a primary cause of cognitive deficits, the pathogenesis of which remains poorly understood. Bruton's tyrosine kinase inhibition (BTKi) is considered a promising strategy to regulate inflammatory responses within the brain, a crucial process that is assumed to drive ischemic demyelination progression. However, the potential role of BTKi in CCH has not been investigated so far. In the present study, we elucidated potential therapeutic roles of BTK in both in vitro hypoxia and in vivo ischemic demyelination model. We found that cerebral hypoperfusion induced white matter injury, cognitive impairments, microglial BTK activation, along with a series of microglia responses associated with inflammation, oxidative stress, mitochondrial dysfunction, and ferroptosis. Tolebrutinib treatment suppressed both the activation of microglia and microglial BTK expression. Meanwhile, microglia-related inflammation and ferroptosis processes were attenuated evidently, contributing to lower levels of disease severity. Taken together, BTKi ameliorated white matter injury and cognitive impairments induced by CCH, possibly via skewing microglia polarization towards anti-inflammatory and homeostatic phenotypes, as well as decreasing microglial oxidative stress damage and ferroptosis, which exhibits promising therapeutic potential in chronic cerebral hypoperfusion-induced demyelination.


Sujet(s)
Agammaglobulinaemia tyrosine kinase , Substance blanche , Animaux , Agammaglobulinaemia tyrosine kinase/antagonistes et inhibiteurs , Agammaglobulinaemia tyrosine kinase/métabolisme , Mâle , Souris , Substance blanche/effets des médicaments et des substances chimiques , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/anatomopathologie , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Encéphalopathie ischémique/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Maladie chronique
3.
Neurosurg Rev ; 47(1): 305, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967704

RÉSUMÉ

The clinical management of aneurysmal subarachnoid hemorrhage (SAH)-associated vasospasm remains a challenge in neurosurgical practice, with its prevention and treatment having a major impact on neurological outcome. While considered a mainstay, nimodipine is burdened by some non-negligible limitations that make it still a suboptimal candidate of pharmacotherapy for SAH. This narrative review aims to provide an update on the pharmacodynamics, pharmacokinetics, overall evidence, and strength of recommendation of nimodipine alternative drugs for aneurysmal SAH-associated vasospasm and delayed cerebral ischemia. A PRISMA literature search was performed in the PubMed/Medline, Web of Science, ClinicalTrials.gov, and PubChem databases using a combination of the MeSH terms "medical therapy," "management," "cerebral vasospasm," "subarachnoid hemorrhage," and "delayed cerebral ischemia." Collected articles were reviewed for typology and relevance prior to final inclusion. A total of 346 articles were initially collected. The identification, screening, eligibility, and inclusion process resulted in the selection of 59 studies. Nicardipine and cilostazol, which have longer half-lives than nimodipine, had robust evidence of efficacy and safety. Eicosapentaenoic acid, dapsone and clazosentan showed a good balance between effectiveness and favorable pharmacokinetics. Combinations between different drug classes have been studied to a very limited extent. Nicardipine, cilostazol, Rho-kinase inhibitors, and clazosentan proved their better pharmacokinetic profiles compared with nimodipine without prejudice with effective and safe neuroprotective role. However, the number of trials conducted is significantly lower than for nimodipine. Aneurysmal SAH-associated vasospasm remains an area of ongoing preclinical and clinical research where the search for new drugs or associations is critical.


Sujet(s)
Encéphalopathie ischémique , Neuroprotecteurs , Nimodipine , Hémorragie meningée , Vasospasme intracrânien , Humains , Hémorragie meningée/complications , Hémorragie meningée/traitement médicamenteux , Vasospasme intracrânien/traitement médicamenteux , Vasospasme intracrânien/étiologie , Nimodipine/usage thérapeutique , Encéphalopathie ischémique/traitement médicamenteux , Neuroprotecteurs/usage thérapeutique , Neuroprotection/effets des médicaments et des substances chimiques , Cilostazol/usage thérapeutique , Nicardipine/usage thérapeutique , Dioxanes/usage thérapeutique , Vasodilatateurs/usage thérapeutique , Pyrimidines/usage thérapeutique , Pyridines , Sulfonamides , Tétrazoles
5.
J Nanobiotechnology ; 22(1): 424, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026255

RÉSUMÉ

Ischemic stroke is a complex, high-mortality disease with multifactorial etiology and pathogenesis. Currently, drug therapy is mainly used treat ischemic stroke in clinic, but there are still some limitations, such as limited blood-brain barrier (BBB) penetration efficiency, a narrow treatment time window and drug side effects. Recent studies have pointed out that drug delivery systems based on polymeric nanocarriers can effectively improve the insufficient treatment for ischemic stroke. They can provide neuronal protection by extending the plasma half-life of drugs, enhancing the drug's permeability to penetrate the BBB, and targeting specific structures and cells. In this review, we classified polymeric nanocarriers used for delivering ischemic stroke drugs and introduced their preparation methods. We also evaluated the feasibility and effectiveness and discussed the existing limitations and prospects of polymeric nanocarriers for ischemic stroke treatment. We hoped that this review could provide a theoretical basis for the future development of nanomedicine delivery systems for the treatment of ischemic stroke.


Sujet(s)
Barrière hémato-encéphalique , Vecteurs de médicaments , Systèmes de délivrance de médicaments , Accident vasculaire cérébral ischémique , Nanoparticules , Polymères , Humains , Polymères/composition chimique , Animaux , Accident vasculaire cérébral ischémique/traitement médicamenteux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments/méthodes , Nanoparticules/composition chimique , Encéphalopathie ischémique/traitement médicamenteux , Nanomédecine/méthodes
6.
J Nanobiotechnology ; 22(1): 393, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965602

RÉSUMÉ

BACKGROUND: The therapeutic strategies for acute ischemic stroke were faced with substantial constraints, emphasizing the necessity to safeguard neuronal cells during cerebral ischemia to reduce neurological impairments and enhance recovery outcomes. Despite its potential as a neuroprotective agent in stroke treatment, Chikusetsu saponin IVa encounters numerous challenges in clinical application. RESULT: Brain-targeted liposomes modified with THRre peptides showed substantial uptake by bEnd. 3 and PC-12 cells and demonstrated the ability to cross an in vitro blood-brain barrier model, subsequently accumulating in PC-12 cells. In vivo, they could significantly accumulate in rat brain. Treatment with C-IVa-LPs-THRre notably reduced the expression of proteins in the P2RX7/NLRP3/Caspase-1 pathway and inflammatory factors. This was evidenced by decreased cerebral infarct size and improved neurological function in MCAO rats. CONCLUSION: The findings indicate that C-IVa-LPs-THRre could serve as a promising strategy for targeting cerebral ischemia. This approach enhances drug concentration in the brain, mitigates pyroptosis, and improves the neuroinflammatory response associated with stroke.


Sujet(s)
Barrière hémato-encéphalique , Accident vasculaire cérébral ischémique , Liposomes , Neuroprotecteurs , Pyroptose , Rat Sprague-Dawley , Saponines , Animaux , Saponines/pharmacologie , Saponines/composition chimique , Pyroptose/effets des médicaments et des substances chimiques , Rats , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Liposomes/composition chimique , Mâle , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Cellules PC12 , Acide oléanolique/pharmacologie , Acide oléanolique/composition chimique , Acide oléanolique/analogues et dérivés , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Peptides/composition chimique , Peptides/pharmacologie , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme
7.
Sci Rep ; 14(1): 15175, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956251

RÉSUMÉ

In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin-eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia-reperfusion injury.


Sujet(s)
Cuivre , Disulfirame , Homéostasie , Inflammation , Souris de lignée C57BL , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Disulfirame/pharmacologie , Souris , Cuivre/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Mâle , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Régulation négative/effets des médicaments et des substances chimiques , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Modèles animaux de maladie humaine , Ferrosulfoprotéines/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Récepteur de type Toll-4/métabolisme
8.
Braz J Med Biol Res ; 57: e13388, 2024.
Article de Anglais | MEDLINE | ID: mdl-38958365

RÉSUMÉ

Jiawei Xinglou Chengqi Granule (JXCG) is an effective herbal medicine for the treatment of ischemic stroke (IS). JXCG has been shown to effectively ameliorate cerebral ischemic symptoms in clinical practice, but the underlying mechanisms are unclear. In this study, we investigated the mechanisms of action of JXCG in the treatment of IS by combining metabolomics with network pharmacology. The chemical composition of JXCG was analyzed using ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). Ultra-high performance liquid chromatography-tandem time-of-flight mass spectrometry (UHPLC-Q-TOF MS) untargeted metabolomics were used to identify differential metabolites within metabolic pathways. Network pharmacology was applied to mine potential targets of JXCG in the treatment of IS. The identified key targets were validated by constructing an integrated network of metabolomics and network pharmacology and by molecular docking using Cytoscape. The effect of JXCG on IS was evaluated in vivo, and the predicted targets and pathways of JXCG in IS therapy were assessed using immunoblotting. Combining metabolomics and network pharmacology, we identified the therapeutic targets of JXCG for IS. Notably, JXCG lessened neuronal damage and reduced cerebral infarct size in rats with IS. Western blot analysis showed that JXCG upregulated PRKCH and downregulated PRKCE and PRKCQ proteins. Our combined network pharmacology and metabolomics findings showed that JXCG may have therapeutic potential in the treatment of IS by targeting multiple factors and pathways.


Sujet(s)
Médicaments issus de plantes chinoises , Accident vasculaire cérébral ischémique , Métabolomique , Pharmacologie des réseaux , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Mâle , Rats , Chromatographie en phase liquide à haute performance , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme
9.
Int Rev Neurobiol ; 177: 95-120, 2024.
Article de Anglais | MEDLINE | ID: mdl-39029992

RÉSUMÉ

The absence of blood flow in cerebral ischemic conditions triggers a multitude of intricate pathophysiological mechanisms, including excitotoxicity, oxidative stress, neuroinflammation, disruption of the blood-brain barrier and white matter disarrangement. Despite numerous experimental studies conducted in preclinical settings, existing treatments for cerebral ischemia (CI), such as mechanical and pharmacological therapies, remain constrained and often entail significant side effects. Therefore, there is an imperative to explore innovative strategies for addressing CI outcomes. Cannabidiol (CBD), the most abundant non-psychotomimetic compound derived from Cannabis sativa, is a pleiotropic substance that interacts with diverse molecular targets and has the potential to influence various pathophysiological processes, thereby contributing to enhanced outcomes in CI. This chapter provides a comprehensive overview of the primary effects of CBD in in vitro and diverse animal models of CI and delves into some of its plausible mechanisms of neuroprotection.


Sujet(s)
Encéphalopathie ischémique , Cannabidiol , Neuroprotecteurs , Cannabidiol/pharmacologie , Cannabidiol/usage thérapeutique , Animaux , Encéphalopathie ischémique/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Humains , Modèles animaux de maladie humaine
11.
Biomolecules ; 14(7)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39062570

RÉSUMÉ

Background: The regulation of divalent metal transporter-1 (DMT1) by insulin has been previously described in Langerhans cells and significant neuroprotection was found by insulin and insulin-like growth factor 1 treatment during experimental cerebral ischemia in acute ischemic stroke patients and in a rat 6-OHDA model of Parkinson's disease, where DMT1 involvement is described. According to the regulation of DMT1, previously described as a target gene of NF-kB in the early phase of post-ischemic neurodegeneration, both in vitro and in vivo, and because insulin controls the NFkB signaling with protection from ischemic cell death in rat cardiomyocytes, we evaluated the role of insulin in relation to DMT1 expression and function during ischemic neurodegeneration. Methods: Insulin neuroprotection is evaluated in differentiated human neuroblastoma cells, SK-N-SH, and in primary mouse cortical neurons exposed to oxygen glucose deprivation (OGD) for 8 h or 3 h, respectively, with or without 300 nM insulin. The insulin neuroprotection during OGD was evaluated in both cellular models in terms of cell death, and in SK-N-SH for DMT1 protein expression and acute ferrous iron treatment, performed in acidic conditions, known to promote the maximum DMT1 uptake as a proton co-transporter; and the transactivation of 1B/DMT1 mouse promoter, already known to be responsive to NF-kB, was analyzed in primary mouse cortical neurons. Results: Insulin neuroprotection during OGD was concomitant to the down-regulation of both DMT1 protein expression and 1B/DMT1 mouse promoter transactivation. We also showed the insulin-dependent protection from cell death after acute ferrous iron treatment. In conclusion, although preliminary, this evaluation highlights the peculiar role of DMT1 as a possible pharmacological target, involved in neuroprotection by insulin during in vitro neuronal ischemia and acute ferrous iron uptake.


Sujet(s)
Transporteurs de cations , Mort cellulaire , Régulation négative , Insuline , Neurones , Animaux , Insuline/métabolisme , Insuline/pharmacologie , Humains , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Souris , Mort cellulaire/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Neuroprotection/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Neuroprotecteurs/pharmacologie , Fer/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Glucose/métabolisme , Composés du fer II/pharmacologie
12.
Neurosurg Rev ; 47(1): 383, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39085665

RÉSUMÉ

This critique discusses neuroprotective strategies for aneurysmal subarachnoid hemorrhage (SAH), excluding Nimodipine, emphasizing alternatives like verapamil, albumin, and cilostazol. While these options show potential, their efficacy lacks robust confirmation from randomized controlled trials (RCTs), relying mainly on observational studies and small trials. The letter underscores the need for comprehensive safety assessments and long-term outcome studies to enhance practical application. Highlighting ongoing trials and emerging therapies like clazosentan and TAK-044, it advocates for future research directions focused on large-scale RCTs and combination therapies, such as cilostazol and Nimodipine, which have demonstrated synergistic benefits in reducing delayed cerebral ischemia (DCI) and improving patient outcomes.


Sujet(s)
Encéphalopathie ischémique , Neuroprotecteurs , Nimodipine , Hémorragie meningée , Vasospasme intracrânien , Humains , Hémorragie meningée/complications , Hémorragie meningée/traitement médicamenteux , Vasospasme intracrânien/traitement médicamenteux , Vasospasme intracrânien/prévention et contrôle , Vasospasme intracrânien/étiologie , Nimodipine/usage thérapeutique , Neuroprotecteurs/usage thérapeutique , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/prévention et contrôle , Neuroprotection/effets des médicaments et des substances chimiques , Cilostazol/usage thérapeutique
13.
Rev Assoc Med Bras (1992) ; 70(6): e20240025, 2024.
Article de Anglais | MEDLINE | ID: mdl-39045961

RÉSUMÉ

OBJECTIVE: Blood-brain barrier is a protective layer that regulates the influx and efflux of biological materials for cerebral tissue. The aim of this study was to investigate the effects of Biochanin A on cerebral histopathology and blood-brain barrier immunohistochemically. METHODS: A total of 24 rats were assigned to three groups: sham, ischemia-reperfusion, and ischemia-reperfusion+Biochanin A. Ischemia-reperfusion was performed by occluding the left carotid artery for 2/24 h. Notably, 20 mg/kg Biochanin A was administered to rats for 7 days after ischemia-reperfusion. Blood was collected for malondialdehyde and total oxidant/antioxidant status analysis. Cerebral tissues were processed for histopathology and further for immunohistochemical analysis. RESULTS: Malondialdehyde content with total oxidant status value was significantly increased and total antioxidant status values were significantly decreased in the ischemia-reperfusion group compared with the sham group. Biochanin A treatment significantly improved scores in the ischemia-reperfusion+Biochanin A group. The normal histological appearance was recorded in the cerebral sections of the sham group. Degenerated neurons and vascular structures with disrupted integrity of the cerebral cortex were observed after ischemia-reperfusion. Biochanin A alleviated the histopathology in the cerebrum in the ischemia-reperfusion+Biochanin A group. Ischemia-reperfusion injury decreased the expression of blood-brain barrier in the ischemia-reperfusion group compared to the sham group. Administration of Biochanin A upregulated the blood-brain barrier immunoreactivity in the cerebrum by restoring blood-brain barrier. CONCLUSION: Cerebral ischemia-reperfusion caused an increase in oxidative stress and pathological lesions in the cerebrum. Biochanin A treatment restored the adverse effects of ischemia-reperfusion injury by restoring blood-brain barrier.


Sujet(s)
Barrière hémato-encéphalique , Génistéine , Malonaldéhyde , Lésion d'ischémie-reperfusion , Animaux , Génistéine/pharmacologie , Génistéine/usage thérapeutique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Mâle , Malonaldéhyde/analyse , Rats , Encéphalopathie ischémique/traitement médicamenteux , Rat Wistar , Antioxydants/pharmacologie , Immunohistochimie , Stress oxydatif/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
15.
Nature ; 631(8022): 826-834, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38987597

RÉSUMÉ

Glutamate is traditionally viewed as the first messenger to activate NMDAR (N-methyl-D-aspartate receptor)-dependent cell death pathways in stroke1,2, but unsuccessful clinical trials with NMDAR antagonists implicate the engagement of other mechanisms3-7. Here we show that glutamate and its structural analogues, including NMDAR antagonist L-AP5 (also known as APV), robustly potentiate currents mediated by acid-sensing ion channels (ASICs) associated with acidosis-induced neurotoxicity in stroke4. Glutamate increases the affinity of ASICs for protons and their open probability, aggravating ischaemic neurotoxicity in both in vitro and in vivo models. Site-directed mutagenesis, structure-based modelling and functional assays reveal a bona fide glutamate-binding cavity in the extracellular domain of ASIC1a. Computational drug screening identified a small molecule, LK-2, that binds to this cavity and abolishes glutamate-dependent potentiation of ASIC currents but spares NMDARs. LK-2 reduces the infarct volume and improves sensorimotor recovery in a mouse model of ischaemic stroke, reminiscent of that seen in mice with Asic1a knockout or knockout of other cation channels4-7. We conclude that glutamate functions as a positive allosteric modulator for ASICs to exacerbate neurotoxicity, and preferential targeting of the glutamate-binding site on ASICs over that on NMDARs may be strategized for developing stroke therapeutics lacking the psychotic side effects of NMDAR antagonists.


Sujet(s)
Canaux ioniques sensibles à l'acidité , Encéphalopathie ischémique , Acide glutamique , Animaux , Femelle , Humains , Mâle , Souris , Amino-2 phosphono-5 valérate/effets indésirables , Amino-2 phosphono-5 valérate/métabolisme , Amino-2 phosphono-5 valérate/pharmacologie , Canaux ioniques sensibles à l'acidité/composition chimique , Canaux ioniques sensibles à l'acidité/déficit , Canaux ioniques sensibles à l'acidité/effets des médicaments et des substances chimiques , Canaux ioniques sensibles à l'acidité/génétique , Canaux ioniques sensibles à l'acidité/métabolisme , Régulation allostérique/effets des médicaments et des substances chimiques , Sites de fixation/génétique , Encéphalopathie ischémique/induit chimiquement , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/anatomopathologie , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Acide glutamique/analogues et dérivés , Acide glutamique/métabolisme , Acide glutamique/pharmacologie , Acide glutamique/toxicité , Souris knockout , Mutagenèse dirigée , Protons , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Récepteurs du N-méthyl-D-aspartate/composition chimique , Récepteurs du N-méthyl-D-aspartate/métabolisme
16.
Int Immunopharmacol ; 138: 112592, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38955024

RÉSUMÉ

Cerebral ischemia-induced systemic inflammation and inflammasome-dependent pyroptotic cell death in ileum, causing serious intestinal injury. Glucocorticoid receptor (GR) mediates the effects of glucocorticoids and participates in inflammation. Escin has corticosteroid-like, neuroprotective, and anti-intestinal dysfunction effects. This study aimed to investigate the effect of Escin on the intestinal barrier injury in rats subjected to middle cerebral artery occlusion (MCAO) and on Caco-2 cells exposed to lipopolysaccharides. The MCAO-caused brain injury was evaluated by assessing neurological function, cerebral infarct volume, and plasma corticosterone (Cort) levels. Intestinal injury was evaluated by observing the histopathological changes, assessing the intestinal barrier function, and determining blood FD4, endotoxin and IL-1ß levels. The levels of the tight-junction proteins such as claudin-1, occludin, and ZO-1, and proteins involved in the GR/p38 MAPK/NF-κB pathway and NLRP3-inflammasome activation were evaluated using western blotting or immunofluorescence. Administration of Escin suppressed the cerebral ischemia-induced increases in Garcia-test scores and infarct volume, alleviated the injury to the intestinal barrier, and decreased the levels of Cort, endotoxin, and IL-1ß. Additionally, Escin upregulated GR and downregulated phospho(p)-p65, p-p38MAPK, NLRP3, GSDMD-N, and cleaved-caspase-1 in the intestine. The effects of Escin could be suppressed by the GR antagonist RU486 or enhanced by the p38 MAPK antagonist SB203580. We revealed details how Escin improves cerebral ischemia-induced intestinal barrier injury by upregulating GR and thereby inhibiting the pyroptosis induced by NF-κB-mediated NLRP3 activation. This study will provide a experimental foundation for the features of glucocorticoid-like activity and the discovery of new clinical application for Escin.


Sujet(s)
Encéphalopathie ischémique , Aescine , Inflammasomes , Pyroptose , Récepteurs aux glucocorticoïdes , Transduction du signal , Animaux , Humains , Mâle , Rats , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Cellules Caco-2 , Modèles animaux de maladie humaine , Aescine/pharmacologie , Aescine/usage thérapeutique , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Infarctus du territoire de l'artère cérébrale moyenne/immunologie , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Intestins/anatomopathologie , Intestins/effets des médicaments et des substances chimiques , Intestins/immunologie , Lipopolysaccharides , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Récepteurs aux glucocorticoïdes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
17.
CNS Neurosci Ther ; 30(7): e14825, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38954749

RÉSUMÉ

AIMS: Ischemic stroke remains a challenge in medical research because of the limited treatment options. Recombinant human tissue plasminogen activator (rtPA) is the primary treatment for recanalization. However, nearly 50% of the patients experience complications that result in ineffective reperfusion. The precise factors contributing to ineffective reperfusion remain unclear; however, recent studies have suggested that immune cells, notably neutrophils, may influence the outcome of rtPA thrombolysis via mechanisms such as the formation of neutrophil extracellular traps. This study aimed to explore the nonthrombolytic effects of rtPA on neutrophils and highlight their contribution to ineffective reperfusion. METHODS: We evaluated the effects of rtPA treatment on middle cerebral artery occlusion in rats. We also assessed neutrophil infiltration and activation after rtPA treatment in vitro and in vivo in a small cohort of patients with massive cerebral ischemia (MCI). RESULTS: rtPA increased neutrophil infiltration into the brain microvessels and worsened blood-brain barrier damage during ischemia. It also increased the neutrophil counts of the patients with MCI. CONCLUSION: Neutrophils play a crucial role in promoting ischemic injury and blood-brain barrier disruption, making them potential therapeutic targets.


Sujet(s)
Fibrinolytiques , Granulocytes neutrophiles , Protéines recombinantes , Activateur tissulaire du plasminogène , Activateur tissulaire du plasminogène/pharmacologie , Activateur tissulaire du plasminogène/usage thérapeutique , Animaux , Humains , Mâle , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Rats , Protéines recombinantes/pharmacologie , Fibrinolytiques/pharmacologie , Fibrinolytiques/usage thérapeutique , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Rat Sprague-Dawley , Sujet âgé , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Femelle , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/immunologie , Modèles animaux de maladie humaine
18.
J Tradit Chin Med ; 44(4): 794-803, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39066540

RÉSUMÉ

OBJECTIVE: To assess the effect and mechanism of Sanhua Tang (, SHT) in treating ischemic stroke (IS) through the "Kaitong Xuanfu" theory by using network pharmacology and animal experiments. METHODS: The active ingredients and targets of SHT and IS were screened by public databases such as Traditional Chinese Medicine systems pharmacology, GeneCards, and online mendelian inheritance in man. Visual network topographies were constructed using R, Cytoscape 3.6.0, AutoDockTools, a user-sponsored molecular visualization system on an open-source foundation, and other software to analyze the correlation between targets and active ingredients. The middle cerebral artery occlusion (MCAO) model was established by operation. Animals were divided into the Sham group, MCAO group (M group), aloe-emodin (AE) group (MCAO rats treated with aloe-emodin), SHT at low dosage (SL group) (MCAO rats treated with SL), SHT at medium dosage (SM group), and SHT at high dosage (SH group). 2,3,5-triphenyl tetrazolium chloride staining was used to detect the volume of cerebral infarction; Nissl staining was used to observe the morphology of neuronal cells; transmission electron microscopy was used to observe the integrity of the blood-brain barrier (BBB); enzyme-linked immunosorbent assay was used to detect the content of interleukin-6 (IL-6), IL-10, tumor necrosis factor α (TNF-α) in serum. Western blot was used to detect the expression of vascular endothelial growth factor A (VEGFA) protein in the cerebral ischemic penumbra. RESULTS: Using network pharmacology and molecular docking validation, four active ingredients (lignan, naringenin, aloe-rhodopsin, and ß-sitosterol), seven target proteins (protein kinase b 1, IL-6, TNF, VEGFA, TP53, jun proto-oncogene, and cysteinyl aspartate specific proteinase 3), and inflammatory signaling pathways were identified. Animal experiments showed that the SH and AE groups had fewer neurological deficits, reduced brain infarct volumes, decreased serum inflammatory factor levels, increased expression of VEGFA protein, and less structural damage to neurons and BBB. CONCLUSION: The present study found that the therapeutic mechanism of SHT against IS may be related to the inhibition of BBB inflammatory damage, which is also the mechanism of "Kaitong Xuanfu." The high-dose group of SHT was relatively effective in regulating inflammatory factors, improving BBB permeability, and protecting neuronal cells from damage.


Sujet(s)
Barrière hémato-encéphalique , Médicaments issus de plantes chinoises , Accident vasculaire cérébral ischémique , Pharmacologie des réseaux , Rat Sprague-Dawley , Animaux , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Rats , Mâle , Humains , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/génétique , Interleukine-6/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/génétique , Neuroprotecteurs/pharmacologie
19.
J Mater Sci Mater Med ; 35(1): 42, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39073469

RÉSUMÉ

Studies have shown that the inhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)was neuroprotective against ischemia/reperfusion(I/R) injury. Bisperoxovanadium (bpV), a derivative of vanadate, is a well-established inhibitor of PTEN. However, its function islimited due to its general inadequacy in penetrating cell membranes. Mxene(Ti3C2Tx) is a novel two-dimensional lamellar nanomaterial with an excellent ability to penetrate the cell membrane. Yet, the effects of this nanomaterial on nervous system diseases have yet to be scrutinized. Here, Mxene(Ti3C2Tx) was used for the first time to carry bpV(HOpic), creating a new nanocomposite Mxene-bpV that was probed in a cerebral I/R injury model. The findings showed that this synthetic Mxene-bpV was adequately stable and can cross the cell membraneeasily. We observed that Mxene-bpV treatment significantly increased the survival rate of oxygen glucose deprivation/reperfusion(OGD/R)--insulted neurons, reduced infarct sizes and promoted the recovery of brain function after mice cerebral I/R injury. Crucially, Mxene-bpV treatment was more therapeutically efficient than bpV(HOpic) treatment alone over the same period. Mechanistically, Mxene-bpV inhibited the enzyme activity of PTEN in vitro and in vivo. It also promoted the expression of phospho-Akt (Ser473) by repressing PTEN and then activated the Akt pathway to boost cell survival. Additionally, in PTEN transgenic mice, Mxene-bpV suppressed I/R-induced inflammatory response by promoting M2 microglial polarization through PTEN inhibition. Collectively, the nanosynthetic Mxene-bpV inhibited PTEN' enzymatic activity by activating Akt pathway and promoting M2 microglial polarization, and finally exerted neuroprotection against cerebral I/R injury.


Sujet(s)
Microglie , Neuroprotecteurs , Phosphohydrolase PTEN , Protéines proto-oncogènes c-akt , Lésion d'ischémie-reperfusion , Transduction du signal , Composés du vanadium , Animaux , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Souris , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/prévention et contrôle , Transduction du signal/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Composés du vanadium/pharmacologie , Composés du vanadium/composition chimique , Phosphohydrolase PTEN/métabolisme , Mâle , Souris de lignée C57BL , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Polarité de la cellule/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Nanocomposites/composition chimique
20.
Drug Des Devel Ther ; 18: 2033-2042, 2024.
Article de Anglais | MEDLINE | ID: mdl-38859883

RÉSUMÉ

Purpose: Odatroltide (LT3001), a novel small synthetic peptide molecule designed to recanalize occluded blood vessels and reduce reperfusion injury, is safe and efficacious in multiple embolic stroke animal models. This study aimed to investigate the safety and tolerability of intravenous administration of odatroltide in patients with acute ischemic stroke within 24 hours of onset. Patients and Methods: Patients with National Institutes of Health Stroke Scale (NIHSS 4-30) who were untreated with intravenous thrombolysis or endovascular thrombectomy were randomized (2:1) to receive a single dose of odatroltide (0.025 mg/kg) or placebo within 24 hours of stroke symptom onset. The primary safety outcome was symptomatic intracranial hemorrhage (sICH) occurrence within 36 hours. Results: Twenty-four patients were enrolled and randomized; of these 16 and 8 received intravenous odatroltide infusion and placebo, respectively. sICH did not occur in both groups, and other safety measures were comparable between the groups. The rate of excellent functional outcome (modified Rankin Scale score, 0-1, at 90 days) was 21% and 14% in the odatroltide and placebo groups, respectively. Furthermore, 47% and 14% of patients in the odatroltide and placebo groups, respectively, showed major neurological improvement (NIHSS improvement ≥4 points from baseline to 30 days). Among the 9 odatroltide-treated patients with baseline NIHSS ≥6, 78% showed major neurological improvement. Conclusion: Compared with placebo, treatment with intravenous odatroltide within 24 hours following onset of ischemic stroke appears to be safe and may be associated with better neurological and functional outcomes. However, the efficacy and safety of odatroltide requires further confirmation in the next phase of clinical trials. Clinical Trial Registration: Clinicaltrials.gov identifier: NCT04091945.


Sujet(s)
Accident vasculaire cérébral ischémique , Humains , Méthode en double aveugle , Mâle , Femelle , Sujet âgé , Accident vasculaire cérébral ischémique/traitement médicamenteux , Adulte d'âge moyen , Facteurs temps , Administration par voie intraveineuse , Perfusions veineuses , Encéphalopathie ischémique/traitement médicamenteux , Sujet âgé de 80 ans ou plus , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE