Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Death Dis ; 15(6): 448, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918408

RÉSUMÉ

Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.


Sujet(s)
Aquaporine-4 , Dexmédétomidine , Système glymphatique , Souris de lignée C57BL , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Sévoflurane , Transduction du signal , Animaux , Dexmédétomidine/pharmacologie , Sévoflurane/pharmacologie , Sévoflurane/effets indésirables , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Souris , Phosphatidylinositol 3-kinases/métabolisme , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Mâle
2.
Exp Neurol ; 378: 114819, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38763355

RÉSUMÉ

BACKGROUND: Both glymphatic system dysfunction and inflammatory response aggravate neurological dysfunction after subarachnoid hemorrhage (SAH). Studies have shown that ß-hydroxybutyrate (BHB) may mitigate painful diabetic neuropathy (PDN) by upregulating SNTA1 expression and reinstating AQP4 polarity. However, the potential of BHB to ameliorate glymphatic system function and inflammatory response in SAH mice remains uncertain. METHODS: The SAH models were constructed by injection of arterial blood into cisterna Magana. Three groups of C57 mice were randomly assigned: Sham, SAH, and BHB. All mice were subjected to neurological function assessment, western blot, immunofluorescence double staining, and RNA-seq. Glymphatic system function was examined with tracer and immunofluorescence double staining, and the differential genes were examined with RNA-seq. In addition, the expression of related inflammation was detected. RESULTS: Compared with the SAH group, BHB reinstated AQP4 polarization by upregulating SNTA1 protein to enhance the glymphatic system. According to RNA-seq, the different genes were primarily connected to microglia activation, astrocytes, and inflammation. Western blot and immunofluorescence further confirmed that the related inflammatory protein expression levels were upregulated. BHB attenuated neuroinflammation after SAH. Ultimately, it can mitigate the neurological deficits in SAH mice. CONCLUSION: The study reveals a novel mechanism that BHB treatment mitigates neurologic impairment in SAH mice. We propose that BHB may play a neuroprotective effect by enhancing glymphatic system function and attenuating neuroinflammatory subarachnoid hemorrhage.


Sujet(s)
Acide 3-hydroxy-butyrique , Système glymphatique , Souris de lignée C57BL , Hémorragie meningée , Animaux , Hémorragie meningée/complications , Hémorragie meningée/métabolisme , Hémorragie meningée/traitement médicamenteux , Souris , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/métabolisme , Mâle , Acide 3-hydroxy-butyrique/pharmacologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/étiologie , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/étiologie , Maladies neuro-inflammatoires/métabolisme
3.
Biomed Pharmacother ; 175: 116739, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38759288

RÉSUMÉ

BACKGROUND: Ketamine, as a non-competitive antagonist of N-methyl-D-aspartate (NMDA) receptors, was originally used in general anesthesia. Epidemiological data show that ketamine has become one of the most commonly abused drugs in China. Ketamine administration might cause cognitive impairment; however, its molecular mechanism remains unclear. The glymphatic system is a lymphoid system that plays a key role in metabolic waste removal and cognitive regulation in the central nervous system. METHODS: Focusing on the glymphatic system, this study evaluated the behavioral performance and circulatory function of the glymphatic system by building a short-term ketamine administration model in mice, and detected the expression levels of the 5-HT2c receptor, ΔFosb, Pten, Akt, and Aqp4 in the hippocampus. Primary astrocytes were cultured to verify the regulatory relationships among related indexes using a 5-HT2c receptor antagonist, a 5-HT2c receptor short interfering RNA (siRNA), and a ΔFosb siRNA. RESULTS: Ketamine administration induced ΔFosb accumulation by increasing 5-HT2c receptor expression in mouse hippocampal astrocytes and primary astrocytes. ΔFosb acted as a transcription factor to recognize the AATGATTAAT bases in the 5' regulatory region of the Aqp4 gene (-1096 bp to -1087 bp), which inhibited Aqp4 expression, thus causing the circulatory dysfunction of the glymphatic system, leading to cognitive impairment. CONCLUSIONS: Although this regulatory mechanism does not involve the Pten/Akt pathway, this study revealed a new mechanism of ketamine-induced cognitive impairment in non-neuronal systems, and provided a theoretical basis for the safety of clinical treatment and the effectiveness of withdrawal.


Sujet(s)
Astrocytes , Dysfonctionnement cognitif , Système glymphatique , Hippocampe , Kétamine , Animaux , Kétamine/pharmacologie , Kétamine/toxicité , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/métabolisme , Souris , Mâle , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Récepteur de la sérotonine de type 5-HT2C/métabolisme , Récepteur de la sérotonine de type 5-HT2C/génétique , Souris de lignée C57BL , Cellules cultivées , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique
4.
Nature ; 623(7989): 992-1000, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37968397

RÉSUMÉ

Cerebral oedema is associated with morbidity and mortality after traumatic brain injury (TBI)1. Noradrenaline levels are increased after TBI2-4, and the amplitude of the increase in noradrenaline predicts both the extent of injury5 and the likelihood of mortality6. Glymphatic impairment is both a feature of and a contributor to brain injury7,8, but its relationship with the injury-associated surge in noradrenaline is unclear. Here we report that acute post-traumatic oedema results from a suppression of glymphatic and lymphatic fluid flow that occurs in response to excessive systemic release of noradrenaline. This post-TBI adrenergic storm was associated with reduced contractility of cervical lymphatic vessels, consistent with diminished return of glymphatic and lymphatic fluid to the systemic circulation. Accordingly, pan-adrenergic receptor inhibition normalized central venous pressure and partly restored glymphatic and cervical lymphatic flow in a mouse model of TBI, and these actions led to substantially reduced brain oedema and improved functional outcomes. Furthermore, post-traumatic inhibition of adrenergic signalling boosted lymphatic export of cellular debris from the traumatic lesion, substantially reducing secondary inflammation and accumulation of phosphorylated tau. These observations suggest that targeting the noradrenergic control of central glymphatic flow may offer a therapeutic approach for treating acute TBI.


Sujet(s)
Oedème cérébral , Lésions traumatiques de l'encéphale , Système glymphatique , Norépinéphrine , Animaux , Souris , Antagonistes adrénergiques/pharmacologie , Antagonistes adrénergiques/usage thérapeutique , Oedème cérébral/complications , Oedème cérébral/traitement médicamenteux , Oedème cérébral/métabolisme , Oedème cérébral/prévention et contrôle , Lésions traumatiques de l'encéphale/complications , Lésions traumatiques de l'encéphale/traitement médicamenteux , Lésions traumatiques de l'encéphale/métabolisme , Modèles animaux de maladie humaine , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/métabolisme , Inflammation/complications , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/prévention et contrôle , Vaisseaux lymphatiques/métabolisme , Norépinéphrine/métabolisme , Phosphorylation , Récepteurs adrénergiques/métabolisme
5.
Ann Neurol ; 90(4): 653-669, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34397111

RÉSUMÉ

OBJECTIVE: Streptococcus pneumoniae is the most common cause of bacterial meningitis, a disease that, despite treatment with antibiotics, still is associated with high mortality and morbidity worldwide. Diffuse brain swelling is a leading cause of morbidity in S pneumoniae meningitis. We hypothesized that neutrophil extracellular traps (NETs) disrupt cerebrospinal fluid (CSF) transport by the glymphatic system and contribute to edema formation in S pneumoniae meningitis. METHODS: We used DNase I treatment to disrupt NETs and then assessed glymphatic function by cisterna magna injections of CSF tracers in a rat model of S pneumoniae meningitis. RESULTS: Our analysis showed that CSF influx into the brain parenchyma, as well as CSF drainage to the cervical lymph nodes, was significantly reduced in the rat model of S pneumoniae meningitis. Degrading NETs by DNase treatment restored glymphatic transport and eliminated the increase in brain weight in the rats. In contrast, first-line antibiotic treatment had no such effect on restoring fluid dynamics. INTERPRETATION: This study suggests that CSF accumulation is responsible for cerebral edema formation and identifies the glymphatic system and NETs as possible new treatment targets in S pneumoniae meningitis. ANN NEUROL 2021;90:653-669.


Sujet(s)
Liquide cérébrospinal/effets des médicaments et des substances chimiques , Désoxyribonucléases/pharmacologie , Pièges extracellulaires/effets des médicaments et des substances chimiques , Méningite à pneumocoques/traitement médicamenteux , Animaux , Encéphale/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Méningite bactérienne/traitement médicamenteux , Méningite à pneumocoques/liquide cérébrospinal , Rat Sprague-Dawley
6.
Mol Neurobiol ; 58(8): 3953-3967, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-33895940

RÉSUMÉ

We have shown that the effects of low-dose ethanol promote the clearance of waste metabolites, such as amyloid-beta (Aß) proteins, from the brain through the perivascular space (PVS). We demonstrated that dilative reactivity of arterial smooth muscle and endothelial cells regulate this clearance. These findings indicate the importance of blood-brain barrier (BBB) transvascular clearance of large size metabolites from the central nervous system (CNS), where the lymphatic clearance system is absent. We next examined the contrasting effects of acute low-dose and chronic moderate ethanol exposure on BBB-associated perivascular clearance. We injected a high molecular weight fluorescent dye into the interstitial space or directly into the cerebrospinal fluid (CSF). Bio-distribution of this tracer was then examined in different brain regions by multiphoton imaging and whole brain tissue section scanning. Ethanol-induced molecular/cellular mechanisms that drive the increase or decrease in movement of the fluorescent tracer were correlated to BBB integrity and arterial vessel reactivity. We found that activation of endothelial nitric oxide synthase (eNOS) under low-dose ethanol conditions with a shift to activation of inducible NOS (iNOS) under chronic high ethanol exposure conditions, which appeared to regulate these contrasting effects. We validated these observations by qualitative and quantitative investigation of eNOS, iNOS, BBB integrity, and perivascular clearance of waste metabolites. We concluded that the effects of low-dose ethanol increased the diffusive movement of waste metabolites via eNOS-derived NO, which increased the arterial endothelial-smooth muscle cell dilative reactivity without affecting BBB integrity, whereas a prolonged induction of iNOS under chronic ethanol exposure conditions caused oxidative damage of the arterial endothelial-smooth muscle layers resulting in cerebral amyloid-like angiopathy. This led to dysfunction of the BBB, dilative reactivity, and impaired waste metabolites movement from the interstitial space or subarachnoid space (SAS) through perivascular clearance.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Endothélium vasculaire/métabolisme , Éthanol/administration et posologie , Système glymphatique/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Transport biologique/physiologie , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Système nerveux central/effets des médicaments et des substances chimiques , Système nerveux central/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Mâle , Nitric oxide synthase type II/métabolisme , Rats , Rat Sprague-Dawley
7.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article de Anglais | MEDLINE | ID: mdl-33525427

RÉSUMÉ

The incidence of brain metastasis has been increasing for 10 years, with poor prognosis, unlike the improvement in survival for extracranial tumor localizations. Since recent advances in molecular biology and the development of specific molecular targets, knowledge of the brain distribution of drugs has become a pharmaceutical challenge. Most anticancer drugs fail to cross the blood-brain barrier. In order to get around this problem and penetrate the brain parenchyma, the use of intrathecal administration has been developed, but the mechanisms governing drug distribution from the cerebrospinal fluid to the brain parenchyma are poorly understood. Thus, in this review we discuss the pharmacokinetics of drugs after intrathecal administration, their penetration of the brain parenchyma and the different systems causing their efflux from the brain to the blood.


Sujet(s)
Antinéoplasiques/pharmacocinétique , Barrière hémato-encéphalique/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Encéphale/effets des médicaments et des substances chimiques , Injections rachidiennes/méthodes , Xénobiotique/pharmacocinétique , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Animaux , Antinéoplasiques/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Liquide cérébrospinal/métabolisme , Expression des gènes , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/métabolisme , Humains , Métastase tumorale , Perméabilité , Récepteur Fc/génétique , Récepteur Fc/métabolisme , Xénobiotique/métabolisme
8.
Fluids Barriers CNS ; 18(1): 1, 2021 Jan 06.
Article de Anglais | MEDLINE | ID: mdl-33407650

RÉSUMÉ

BACKGROUND: Large differences in glymphatic system transport-similar in magnitude to those of the sleep/wake cycle-have been observed during anesthesia with dexmedetomidine supplemented with low dose isoflurane (DEXM-I) in comparison to isoflurane (ISO). However, the biophysical and bioenergetic tissue status underlying glymphatic transport differences between anesthetics remains undefined. To further understand biophysical characteristics underlying these differences we investigated volume status across cerebral tissue compartments, water diffusivity, and T2* values in rats anesthetized with DEXM-I in comparison to ISO. METHODS: Using a crossover study design, a group of 12 Sprague Dawley female rats underwent repetitive magnetic resonance imaging (MRI) under ISO and DEXM-I. Physiological parameters were continuously measured. MRI included a proton density weighted (PDW) scan to investigate cerebrospinal fluid (CSF) and parenchymal volumetric changes, a multigradient echo scan (MGE) to calculate T2* maps as a measure of 'bioenergetics', and a diffusion scan to quantify the apparent diffusion coefficient (ADC). RESULTS: The heart rate was lower with DEXM-I in comparison to ISO, but all other physiological variables were similar across scans and groups. The PDW images revealed a 1% parenchymal volume increase with ISO compared to DEXM-I comprising multiple focal tissue areas scattered across the forebrain. In contrast, with DEXM-I the CSF compartment was enlarged by ~ 6% in comparison to ISO at the level of the basal cisterns and peri-arterial conduits which are main CSF influx routes for glymphatic transport. The T2* maps showed brain-wide increases in T2* in ISO compared to DEXM-I rats. Diffusion-weighted images yielded no significant differences in ADCs across the two anesthesia groups. CONCLUSIONS: We demonstrated CSF volume expansion with DEXM-I (in comparison to ISO) and parenchymal (GM) expansion with ISO (in comparison to DEXM-I), which may explain the differences in glymphatic transport. The T2* changes in ISO are suggestive of an increased bioenergetic state associated with excess cellular firing/bursting when compared to DEXM-I.


Sujet(s)
Anesthésiques/pharmacologie , Liquide cérébrospinal/effets des médicaments et des substances chimiques , Dexmédétomidine/pharmacologie , Transferts liquidiens/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Substance grise/effets des médicaments et des substances chimiques , Isoflurane/pharmacologie , Animaux , Liquide cérébrospinal/imagerie diagnostique , Études croisées , Femelle , Système glymphatique/imagerie diagnostique , Substance grise/imagerie diagnostique , Imagerie par résonance magnétique , Rats , Rat Sprague-Dawley
9.
J Neuroinflammation ; 18(1): 34, 2021 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-33514389

RÉSUMÉ

BACKGROUND: The exchange of cerebrospinal (CSF) and interstitial fluid is believed to be vital for waste clearance in the brain. The sleep-dependent glymphatic system, which is comprised of perivascular flow of CSF and is largely dependent on arterial pulsatility and astrocytic aquaporin-4 (AQP4) expression, facilitates much of this brain clearance. During the last decade, several observations have indicated that impaired glymphatic function goes hand in hand with neurodegenerative diseases. Since pathologies of the brain carry inflammatory components, we wanted to know how acute inflammation, e.g., with lipopolysaccharide (LPS) injections, would affect the glymphatic system. In this study, we aim to measure the effect of LPS on perivascular CSF distribution as a measure of glymphatic function. METHODS: Three hours after injection of LPS (1 mg/kg i.p.), C57bl/6 mice were (1) imaged for two CSF tracers, injected into cisterna magna, (2) transcardially perfused with buffer, or (3) used for physiological readouts. Tracer flow was imaged using a low magnification microscope on fixed brains, as well as using vibratome-cut slices for measuring tracer penetration in the brain. Cytokines, glial, and BBB-permeability markers were measured with ELISAs, Western blots, and immunohistochemistry. Cerebral blood flow was approximated using laser Doppler flowmetry, respiration and heart rate with a surgical monitor, and AQP4-polarization was quantified using confocal microscopy of immunolabeled brain sections. RESULTS: LPS-injections significantly lowered perivascular CSF tracer flow and penetration into the parenchyma. No differences in AQP4 polarization, cytokines, astroglial and BBB markers, cerebral blood flow, or respiration were detected in LPS-injected mice, although LPS did elevate cortical Iba1+ area and heart rate. CONCLUSIONS: This study reports another physiological response after acute exposure to the bacterial endotoxin LPS, namely the statistically significant decrease in perivascular distribution of CSF. These observations may benefit our understanding of the role of systemic inflammation in brain clearance.


Sujet(s)
Liquide cérébrospinal/métabolisme , Liquide extracellulaire/métabolisme , Système glymphatique/métabolisme , Lipopolysaccharides/toxicité , Animaux , Liquide cérébrospinal/composition chimique , Liquide cérébrospinal/effets des médicaments et des substances chimiques , Circulation cérébrovasculaire/effets des médicaments et des substances chimiques , Circulation cérébrovasculaire/physiologie , Liquide extracellulaire/composition chimique , Liquide extracellulaire/effets des médicaments et des substances chimiques , Colorants fluorescents/administration et posologie , Colorants fluorescents/métabolisme , Système glymphatique/composition chimique , Système glymphatique/effets des médicaments et des substances chimiques , Fluxmétrie laser Doppler/méthodes , Mâle , Souris , Souris de lignée C57BL
10.
Comb Chem High Throughput Screen ; 24(7): 1083-1092, 2021.
Article de Anglais | MEDLINE | ID: mdl-32875981

RÉSUMÉ

Alzheimer's disease is an age-related neurodegenerative disease. The factors causing Alzheimer's disease are numerous. Research on humans and rodent models predicted various causative factors involved in Alzheimer's disease progression. Among them, neuroinflammation, oxidative stress, and apoptosis play a major role because of the accumulation of extracellular amyloid-beta peptides. Here, the clearance of amyloid beta-peptide plays a major role because of the imbalance in the production and clearance of the amyloid beta-peptide. Additionally, neuroinflammation by microglia, astrocytes, cytokines, chemokines, and the complement system also has a major role in Alzheimer's disease. The physiological clearance pathways involved in amyloid beta-peptide are glymphatic, vascular, and immune pathways. Amyloid precursor protein, low-density lipoprotein receptor-related protein 1, receptor for the advanced glycation end product, apolipoprotein E, clusterin, aquaporin 4, auto-antibodies, complement system, cytokines, and microglia are involved in amyloid beta-peptide clearance pathways across the blood-brain barrier. The plaque formation in the brain by alternative splicing of amyloid precursor protein and production of misfolded protein results in amyloid-beta agglomeration. This insoluble amyloid-beta leads to a neurodegenerative cascade and neuronal cell death occurs. Studies had shown that disturbed sleep may be a risk factor for dementia and cognitive decline. In this review, the therapeutic targets for Alzheimer'sdisease via focusing on pathways for amyloid-beta clearance are discussed.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Système glymphatique/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Maladie d'Alzheimer/immunologie , Peptides bêta-amyloïdes/immunologie , Animaux , Système glymphatique/immunologie , Humains
11.
J Mol Neurosci ; 71(6): 1266-1274, 2021 Jun.
Article de Anglais | MEDLINE | ID: mdl-33188502

RÉSUMÉ

Amyloid-ß (Aß) deposit in the parenchyma is a major characteristic in Alzheimer's disease (AD), and the impaired glymphatic clearance contributes to the Aß accumulation. It was reported that L-3-n-butylphthalide (NBP) showed the potential neuroprotective effect in the rodent models of AD. The effects of NBP on the glymphatic system were explored in this study. In the wild-type mice, both CSF tracer influx and perivascular drainage increased after NBP treatment compared with vehicle treatment. Moreover, NBP promoted the perivascular drainage of Aß via increased cerebral pulsation, which could be inhibited by propranolol. Then, we studied the potential of 3-month NBP treatment on Aß deposits in 8-month-old APP/PS1 transgenic mice. NBP daily treatments remarkably improved cognitive behavior in Morris water maze. Furthermore, NBP could reduce Aß deposition and decrease parenchymal Aß levels. In addition, NBP markedly improved the perivascular AQP4 localization. Our results indicated that NBP could enhance the glymphatic clearance and reduce parenchymal Aß deposit in the APP/PS1 mice, suggesting that it may have potential in the treatment of AD.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/métabolisme , Benzofuranes/pharmacologie , Système glymphatique/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Animaux , Benzofuranes/usage thérapeutique , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Système glymphatique/métabolisme , Mâle , Apprentissage du labyrinthe , Souris , Souris de lignée C57BL , Neuroprotecteurs/usage thérapeutique
12.
Biomed Pharmacother ; 132: 110887, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-33254429

RÉSUMÉ

Alzheimer's disease (AD) is an age-related neurodegenerative disease with multiple predisposing factors and complicated pathogenesis. Aß peptide is one of the most important pathogenic factors in the etiology of AD. Accumulating evidence indicates that the imbalance of Aß production and Aß clearance in the brain of AD patients leads to Aß deposition and neurotoxic Aß oligomer formation. Melatonin shows a potent neuroprotective effect and can prevent or slow down the progression of AD, supporting the view that melatonin is a potential therapeutic molecule for AD. Melatonin modulates the regulatory network of secretase expression and affects the function of secretase, thereby inhibiting amyloidogenic APP processing and Aß production. Additionally, melatonin ameliorates Aß-induced neurotoxicity and probably promotes Aß clearance through glymphatic-lymphatic drainage, BBB transportation and degradation pathways. In this review, we summarize and discuss the role of melatonin against Aß-dependent AD pathogenesis. We explore the potential cellular and molecular mechanisms of melatonin on Aß production and assembly, Aß clearance, Aß neurotoxicity and circadian cycle disruption. We summarize multiple clinical trials of melatonin treatment in AD patients, showing that melatonin has a promising effect on improving sleep quality and cognitive function. This review aims to stimulate further research on melatonin as a potential therapeutic agent for AD.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Encéphale/métabolisme , Système glymphatique/métabolisme , Mélatonine/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Animaux , Encéphale/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Humains , Mélatonine/administration et posologie
13.
Sci Rep ; 10(1): 16144, 2020 09 30.
Article de Anglais | MEDLINE | ID: mdl-32999351

RÉSUMÉ

Drainage of parenchymal waste through the lymphatic system maintains brain homeostasis. Age-related changes of glymphatic-lymphatic clearance lead to the accumulation beta-amyloid (Aß) in dementia models. In this study, focused ultrasound treatment in combination with microbubbles (FUS-MB) improved Aß drainage in early dementia model mice, 5XFAD. FUS-MB enhanced solute Aß clearance from brain, but not plaques, to cerebrospinal fluid (CSF) space and then deep cervical lymph node (dCLN). dCLN ligation exaggerated memory impairment and progress of plaque formation and also the beneficial effects of FUS-MB upon Aß removal through CSF-lymphatic routes. In this ligation model, FUS-MB improved memory despite accumulation of Aß in CSF. In conclusion, FUS-MB enhances glymphatic-lymphatic clearance of Aß mainly by increasing brain-to-CSF Aß drainage. We suggest that FUS-MB can delay dementia progress in early period and benefits of FUS-MB depend on the effect of Aß disposal through CSF-lymphatics.


Sujet(s)
Maladie d'Alzheimer/thérapie , Système glymphatique/effets des médicaments et des substances chimiques , Microbulles/usage thérapeutique , Maladie d'Alzheimer/immunologie , Peptides bêta-amyloïdes/métabolisme , Animaux , Encéphale/métabolisme , Modèles animaux de maladie humaine , Système glymphatique/métabolisme , Humains , Système lymphatique/métabolisme , Mâle , Souris , Souris transgéniques , Tissu parenchymateux , Plaque amyloïde/anatomopathologie , Ultrasonothérapie/méthodes
14.
Neurotherapeutics ; 17(4): 1954-1972, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32918234

RÉSUMÉ

Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.


Sujet(s)
Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Oedème cérébral/traitement médicamenteux , Système glymphatique/effets des médicaments et des substances chimiques , Polypeptide activateur de l'adénylcyclase hypophysaire/administration et posologie , Hémorragie meningée/traitement médicamenteux , Animaux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Oedème cérébral/métabolisme , Oedème cérébral/anatomopathologie , Système glymphatique/métabolisme , Système glymphatique/anatomopathologie , Mâle , Rats , Rat Sprague-Dawley , Hémorragie meningée/métabolisme , Hémorragie meningée/anatomopathologie
15.
Mol Pharm ; 17(11): 4067-4076, 2020 11 02.
Article de Anglais | MEDLINE | ID: mdl-32955898

RÉSUMÉ

Direct drug delivery from nose to brain has drawn much attention as an effective strategy for the treatment of central nervous system diseases. After intranasal administration, drug molecules can be directly delivered from the nose to the brain. However, the detailed mechanism for this direct delivery to the brain has not been elucidated. In the present study, the effect of the activation of the cerebral fluid circulation (the glymphatic system) on the efficacy of direct delivery from nose to brain was investigated. Because the glymphatic system is activated by some anesthetic regimens, the differences in brain delivery and the pharmacokinetics under anesthetic and conscious conditions were compared in rats. Under urethane anesthesia, direct delivery from the nose to the brain was facilitated, whereas the brain uptake from the systemic circulation via the blood-brain barrier was decreased. In addition, both the brain uptake of caffeine injected into the subarachnoid cerebrospinal fluid (CSF) and the extracerebral clearance of caffeine after intrastriatal injection were enhanced under anesthesia. For intranasal administration, caffeine was transported directly from the nose to the CSF and then delivered into the brain parenchyma by the CSF circulation. The results obtained in the present study clarified that the direct delivery from nose to brain could be facilitated by anesthesia. These findings suggest that fluid circulation in the brain can contribute to a wider cerebral distribution of the drug after direct delivery from nose to brain.


Sujet(s)
Administration par voie nasale/méthodes , Barrière hémato-encéphalique/métabolisme , Caféine/administration et posologie , Caféine/sang , Liquide cérébrospinal/métabolisme , Système glymphatique/métabolisme , Muqueuse nasale/métabolisme , Anesthésie/méthodes , Animaux , Transport biologique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Caféine/liquide cérébrospinal , Caféine/pharmacocinétique , Liquide cérébrospinal/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Mâle , Muqueuse nasale/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Résultat thérapeutique
16.
J Control Release ; 304: 29-38, 2019 06 28.
Article de Anglais | MEDLINE | ID: mdl-31067483

RÉSUMÉ

Drug delivery to the central nervous system remains a major problem due to biological barriers. The blood-brain-barrier can be bypassed by administering drugs intrathecally directly to the cerebrospinal fluid (CSF). The glymphatic system, a network of perivascular spaces promoting fluid exchange between CSF and interstitial space, could be utilized to enhance convective drug delivery from the CSF to the parenchyma. Glymphatic flow is highest during sleep and anesthesia regimens that induce a slow-wave sleep-like state. Here, using mass spectrometry and fluorescent imaging techniques, we show that the clinically used α2-adrenergic agonist dexmedetomidine that enhances EEG slow-wave activity, increases brain and spinal cord drug exposure of intrathecally administered drugs in mice and rats. Using oxycodone, naloxone, and an IgG-sized antibody as relevant model drugs we demonstrate that modulation of glymphatic flow has a distinct impact on the distribution of intrathecally administered therapeutics. These findings can be exploited in the clinic to improve the efficacy and safety of intrathecally administered therapeutics.


Sujet(s)
Encéphale/métabolisme , Dexmédétomidine/administration et posologie , Systèmes de délivrance de médicaments , Système glymphatique/effets des médicaments et des substances chimiques , Agonistes des récepteurs alpha-2 adrénergiques/administration et posologie , Agonistes des récepteurs alpha-2 adrénergiques/pharmacologie , Animaux , Anticorps/administration et posologie , Anticorps/métabolisme , Dexmédétomidine/pharmacologie , Système glymphatique/métabolisme , Injections rachidiennes , Mâle , Souris , Souris de lignée C57BL , Naloxone/administration et posologie , Naloxone/pharmacocinétique , Oxycodone/administration et posologie , Oxycodone/pharmacocinétique , Rats , Rat Sprague-Dawley , Distribution tissulaire
17.
Psychopharmacology (Berl) ; 236(4): 1367-1384, 2019 Apr.
Article de Anglais | MEDLINE | ID: mdl-30607477

RÉSUMÉ

BACKGROUND: The glymphatic system has recently been proposed to function as a brain-wide macroscopic system for the clearance of potentially harmful molecules, such as amyloid beta (e.g., Aß), from the brain parenchyma. Previous literatures have established that the glymphatic function is dramatically suppressed by aging, traumatic brain injury, and some diseases. However, the effect of chronic stress on the glymphatic function and its underlying mechanism remains largely unknown. METHODS: Adult mice were randomly divided into four groups: chronic unpredictable mild stress (CUMS)-treated group, CUMS simultaneously treated with mifepristone (MFP) group, dexamethasone (DEX)-treated group, and control group. Stress response was observed by assessing the change of body weight, plasma corticosterone level, and behavior tests. The level of Aß42 in cerebral tissue was assessed by ELISA. The glymphatic function was determined by using fluorescence tracer injection. The expression and localization of aquaporin-4 (AQP4) were evaluated by immunohistochemistry and western blot. The transcription level of AQP4 and anchoring molecules was evaluated by real-time PCR. FINDINGS: Compared with control group, CUMS-treated mice exhibited the impairment of global glymphatic function especially in the anterior brain. This change was accompanied by the decreased expression and polarization of AQP4, reduced transcription of AQP4, agrin, laminin, and dystroglycan in the anterior cortex. Similarly, the glucocorticoid receptor (GR) agonist DEX exposure could reduce the glymphatic function and AQP4 expression. Moreover, the GR antagonist MFP treatment could significantly rescue the glymphatic function and reverse the expression and polarization of AQP4 impaired by CUMS. CONCLUSION: Chronic stress could impair the AQP4-mediated glymphatic transport in the brain through glucocorticoid signaling. Our results also suggest that GR antagonist could be beneficial to rescue the glymphatic function suppressed by chronic stress.


Sujet(s)
Aquaporine-4/métabolisme , Glucocorticoïdes/métabolisme , Système glymphatique/métabolisme , Transduction du signal/physiologie , Stress psychologique/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Dexaméthasone/toxicité , Système glymphatique/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL , Répartition aléatoire , Récepteurs aux glucocorticoïdes/agonistes , Récepteurs aux glucocorticoïdes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Stress psychologique/induit chimiquement , Stress psychologique/psychologie
18.
JCI Insight ; 3(20)2018 10 18.
Article de Anglais | MEDLINE | ID: mdl-30333324

RÉSUMÉ

Despite the initial promise of immunotherapy for CNS disease, multiple recent clinical trials have failed. This may be due in part to characteristically low penetration of antibodies to cerebrospinal fluid (CSF) and brain parenchyma, resulting in poor target engagement. We here utilized transcranial macroscopic imaging to noninvasively evaluate in vivo delivery pathways of CSF fluorescent tracers. Tracers in CSF proved to be distributed through a brain-wide network of periarterial spaces, previously denoted as the glymphatic system. CSF tracer entry was enhanced approximately 3-fold by increasing plasma osmolality without disruption of the blood-brain barrier. Further, plasma hyperosmolality overrode the inhibition of glymphatic transport that characterizes the awake state and reversed glymphatic suppression in a mouse model of Alzheimer's disease. Plasma hyperosmolality enhanced the delivery of an amyloid-ß (Aß) antibody, obtaining a 5-fold increase in antibody binding to Aß plaques. Thus, manipulation of glymphatic activity may represent a novel strategy for improving penetration of therapeutic antibodies to the CNS.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Liquide cérébrospinal/métabolisme , Système glymphatique/métabolisme , Immunothérapie/méthodes , Maladie d'Alzheimer/anatomopathologie , Animaux , Barrière hémato-encéphalique/métabolisme , Modèles animaux de maladie humaine , Colorants fluorescents/administration et posologie , Colorants fluorescents/pharmacocinétique , Système glymphatique/imagerie diagnostique , Système glymphatique/effets des médicaments et des substances chimiques , Humains , Facteurs immunologiques/administration et posologie , Facteurs immunologiques/pharmacocinétique , Injections ventriculaires , Microscopie intravitale , Mâle , Mannitol/administration et posologie , Souris , Imagerie optique , Concentration osmolaire , Perméabilité/effets des médicaments et des substances chimiques , Plasma sanguin/composition chimique , Plasma sanguin/effets des médicaments et des substances chimiques , Solution saline hypertonique/administration et posologie
19.
Trends Mol Med ; 24(10): 838-855, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30100517

RÉSUMÉ

The lack of effective treatment options for chronic neurological conditions, such as multiple sclerosis (MS), highlights the need to re-evaluate disease pathophysiology in the process of identifying novel therapeutic targets. The persistent activation of mononuclear phagocytes (MPs) is one of the major drivers of neurodegeneration and it sustains central nervous system (CNS) damage. Mitochondrial metabolism influences the activity of MPs, and the metabolites that they produce have key signalling roles in inflammation. However, how changes in immune cell metabolism sustain a chronic state of neuroinflammation is not fully understood. Novel molecular and cellular therapies for chronic neuroinflammation should be developed to target mitochondrial metabolism in innate immune cells to prevent secondary neurological damage and the accumulation of irreversible disability in patients.


Sujet(s)
Système nerveux central/métabolisme , Mitochondries/métabolisme , Thérapie moléculaire ciblée/méthodes , Sclérose en plaques chronique progressive/métabolisme , Maladies neurodégénératives/métabolisme , Système nerveux central/effets des médicaments et des substances chimiques , Système nerveux central/immunologie , Système nerveux central/anatomopathologie , Crotonates/usage thérapeutique , Fumarate de diméthyle/usage thérapeutique , Transport d'électrons/effets des médicaments et des substances chimiques , Système glymphatique/effets des médicaments et des substances chimiques , Système glymphatique/immunologie , Système glymphatique/métabolisme , Système glymphatique/anatomopathologie , Humains , Hydroxy-butyrates , Immunité innée , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Voies et réseaux métaboliques/immunologie , Microglie/effets des médicaments et des substances chimiques , Microglie/immunologie , Microglie/métabolisme , Microglie/anatomopathologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/immunologie , Mitochondries/anatomopathologie , Système phagocytaire mononucléé/effets des médicaments et des substances chimiques , Système phagocytaire mononucléé/immunologie , Système phagocytaire mononucléé/métabolisme , Système phagocytaire mononucléé/anatomopathologie , Sclérose en plaques chronique progressive/immunologie , Sclérose en plaques chronique progressive/anatomopathologie , Sclérose en plaques chronique progressive/thérapie , Maladies neurodégénératives/immunologie , Maladies neurodégénératives/anatomopathologie , Maladies neurodégénératives/thérapie , Nitriles , Transduction du signal , Transplantation de cellules souches/méthodes , Toluidines/usage thérapeutique
20.
Sci Rep ; 8(1): 2246, 2018 02 02.
Article de Anglais | MEDLINE | ID: mdl-29396480

RÉSUMÉ

Prolonged intake of excessive amounts of ethanol is known to have adverse effects on the central nervous system (CNS). Here we investigated the effects of acute and chronic ethanol exposure and withdrawal from chronic ethanol exposure on glymphatic function, which is a brain-wide metabolite clearance system connected to the peripheral lymphatic system. Acute and chronic exposure to 1.5 g/kg (binge level) ethanol dramatically suppressed glymphatic function in awake mice. Chronic exposure to 1.5 g/kg ethanol increased GFAP expression and induced mislocation of the astrocyte-specific water channel aquaporin 4 (AQP4), but decreased the levels of several cytokines. Surprisingly, glymphatic function increased in mice treated with 0.5 g/kg (low dose) ethanol following acute exposure, as well as after one month of chronic exposure. Low doses of chronic ethanol intake were associated with a significant decrease in GFAP expression, with little change in the cytokine profile compared with the saline group. These observations suggest that ethanol has a J-shaped effect on the glymphatic system whereby low doses of ethanol increase glymphatic function. Conversely, chronic 1.5 g/kg ethanol intake induced reactive gliosis and perturbed glymphatic function, which possibly may contribute to the higher risk of dementia observed in heavy drinkers.


Sujet(s)
Consommation d'alcool/effets indésirables , Alcoolisme/anatomopathologie , Éthanol/administration et posologie , Éthanol/pharmacologie , Système glymphatique/effets des médicaments et des substances chimiques , Animaux , Aquaporine-4/effets des médicaments et des substances chimiques , Cytokines/sang , Démence/induit chimiquement , Gliose/induit chimiquement , Mâle , Souris , Souris de lignée C57BL , Sommeil/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...