Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 76
Filtrer
1.
Acta Pharmacol Sin ; 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112771

RÉSUMÉ

Esculetin (ESC) is a coumarin-derived phytochemical prevalent in traditional Chinese medicine that exhibits anti-acute ischemic stroke activities. Our previous studies demonstrate that CKLF1 is a potential anti-stroke target for coumarin-derived compound. In this study we investigated whether CKLF1 was involved in the neuroprotective effects of ESC against photothrombotic stroke in mice. The mice were treated with ESC (20, 40 or 80 mg·kg-1·d-1, i.g.) for two weeks. The therapeutic effect of ESC was assessed using MRI, neurological function evaluation, and a range of behavioral tests on D1, 3, 7 and 14 of ESC administration. We showed that oral administration of ESC dose-dependently reduced the cerebral infarction volume within one week after stroke, improved behavioral performance, and alleviated neuropathological damage within two weeks. Functional MRI revealed that ESC significantly enhanced the abnormal low-frequency fluctuation (ALFF) value of the motor cortex and promoted functional connectivity between the supplementary motor area (SMA) and multiple brain regions. We demonstrated that ESC significantly reduced the protein levels of CKLF1 and CCR5, as well as the CKLF1/CCR5 protein complex in the peri-infarcted area. We showed that ESC (0.1-10 µM) dose-dependently blocked CKLF1-induced chemotactic movement of neutrophils in the Transwell assay, reducing the interaction of CKLF1/CCR5 on the surface of neutrophils, thereby reducing neutrophil infiltration, and decreasing the expression of ICAM-1, VCAM-1 and MMP-9 in the peri-infarct tissue. Knockout of CKLF1 reduced brain infarction volume and motor dysfunction after stroke but also negated the anti-stroke efficacy and neutrophil infiltration of ESC. These results suggest that the efficacy of ESC in promoting post-stroke neural repair depends on its inhibition on CKLF1-mediated neutrophil infiltration, which offering novel perspectives for elucidating the therapeutic properties of coumarins.

7.
Pharmacol Biochem Behav ; 239: 173757, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38574898

RÉSUMÉ

Depression is a major chronic mental illness worldwide, characterized by anhedonia and pessimism. Exposed to the same stressful stimuli, some people behave normally, while others exhibit negative behaviors and psychology. The exact molecular mechanisms linking stress-induced depressive susceptibility and resilience remain unclear. Connexin 43 (Cx43) forms gap junction channels between the astrocytes, acting as a crucial role in the pathogenesis of depression. Cx43 dysfunction could lead to depressive behaviors, and depression down-regulates the expression of Cx43 in the prefrontal cortex (PFC). Besides, accumulating evidence indicates that inflammation is one of the most common pathological features of the central nervous system dysfunction. However, the roles of Cx43 and peripheral inflammation in stress-susceptible and stress-resilient individuals have rarely been investigated. Thus, animals were classified into the chronic unpredictable stress (CUS)-susceptible group and the CUS-resilient group based on the performance of behavioral tests following the CUS protocol in this study. The protein expression of Cx43 in the PFC, the Cx43 functional changes in the PFC, and the expression levels including interleukin (IL)-1ß, tumor necrosis factor-α, IL-6, IL-2, IL-10, and IL-18 in the peripheral serum were detected. Here, we found that stress exposure triggered a significant reduction in Cx43 protein expression in the CUS-susceptible mice but not in the CUS-resilient mice accompanied by various Cx43 phosphorylation expression and the changes of inflammatory signals. Stress resilience is associated with Cx43 in the PFC and fluctuation in inflammatory signaling, showing that therapeutic targeting of these pathways might promote stress resilience.


Sujet(s)
Connexine 43 , Inflammation , Cortex préfrontal , Stress psychologique , Animaux , Cortex préfrontal/métabolisme , Connexine 43/métabolisme , Souris , Stress psychologique/métabolisme , Mâle , Inflammation/métabolisme , Résilience psychologique , Souris de lignée C57BL , Dépression/métabolisme , Cytokines/métabolisme , Prédisposition aux maladies , Comportement animal
8.
Zhongguo Zhong Yao Za Zhi ; 49(1): 55-61, 2024 Jan.
Article de Chinois | MEDLINE | ID: mdl-38403338

RÉSUMÉ

The theory of kidney storing essence storage, an important part of the basic theory of traditional Chinese medicine(TCM), comes from the Chapter 9 Discussion on Six-Plus-Six System and the Manifestations of the Viscera in the Plain Questions, which says that "the kidney manages closure and is the root of storage and the house of Jing(Essence)". According to this theory, essence is the fundamental substance of human life activities and it is closely related to the growth and development of the human body. Alzheimer's disease(AD) is one of the common neurodegenerative diseases, with the main pathological features of Aß deposition and Tau phosphorylation, which activate neurotoxic reactions and eventually lead to neuronal dysfunction and cell death, severely impairing the patient's cognitive and memory functions. Although research results have been achieved in the TCM treatment of AD, the complex pathogenesis of AD makes it difficult to develop the drugs capable of curing AD. The stem cell therapy is an important method to promote self-repair and regeneration, and bone marrow mesenchymal stem cells(BMSCs) as adult stem cells have the ability of multi-directional differentiation. By reviewing the relevant literature, this paper discusses the association between BMSCs and the TCM theory of kidney storing essence, and expounds the material basis of this theory from the perspective of molecular biology. Studies have shown that TCM with the effect of tonifying the kidney in the treatment of AD are associated with BMSCs. Exosomes produced by such cells are one of the main substances affecting AD. Exosomes containing nucleic acids, proteins, and lipids can participate in intercellular communication, regulate cell function, and affect AD by reducing Aß deposition, inhibiting Tau protein phosphorylation and neuroinflammation, and promoting neuronal regeneration. Therefore, discussing the prevention and treatment of exosomes and AD based on the theory of kidney storing essence will provide a new research idea for the TCM treatment of AD.


Sujet(s)
Maladie d'Alzheimer , Exosomes , Adulte , Humains , Maladie d'Alzheimer/prévention et contrôle , Maladie d'Alzheimer/traitement médicamenteux , Exosomes/métabolisme , Exosomes/anatomopathologie , Rein/anatomopathologie , Médecine traditionnelle chinoise , Neurones
9.
Brain Res Bull ; 207: 110871, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38211740

RÉSUMÉ

CONTEXT: Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, has been shown to exhibit anti-depressive effects in clinical trials. However, the direct mechanism underlying its effect on neuroinflammation remains unclear. Neuroinflammatory reaction from astrocytes leads to depression, and our previous study found that gap junction disorder between astrocytes aggravated neuroinflammatory reaction in depressed mice. OBJECTIVE: To investigate the potential mechanism of celecoxib's effects on astrocytic gap junctions during the central nervous inflammation-induced depression. MATERIALS & METHODS: Stereotaxic injection of lipopolysaccharide (LPS) into the prefrontal cortex (PFC) to establish a model of major depressive disorder (MDD). Celecoxib was administrated into PFC 15 min after LPS injection. The depressive performance was tested by tail suspension test and forced swimming test, and the levels of proinflammation cytokines were determined at mRNA and protein levels. Resting-state functional connection (rsFC) was employed to assess changes in the default mode network (DMN). Additionally, astrocytic gap junctions were also determined by lucifer yellow (LY) diffusion and transmission electron microscope (TEM), and the expression of connexin 43 (Cx43) was measured by western blotting, quantitative polymerase chain reaction, and immunofluorescence. RESULTS: LPS injection induced significant depressive performance, which was ameliorated by celecoxib treatment. Celecoxib also improved rsFC in the DMN. Furthermore, celecoxib improved astrocytic gap junctions as evidenced by increased LY diffusion, shortened gap junction width, and normalized levels of phosphorylated Cx43. Celecoxib also blocked the phosphorylation of p65, and inhibition of p65 abolished the improvement of Cx43. DISCUSSION & CONCLUSION: Anti-depressive effects of celecoxib are mediated, at least in part, by the inhibition of nuclear factor- kappa B (NF-κB) and the subsequent improvement of astrocytic gap junction function.


Sujet(s)
Trouble dépressif majeur , Facteur de transcription NF-kappa B , Animaux , Souris , Célécoxib/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Connexine 43/métabolisme , Astrocytes/métabolisme , Trouble dépressif majeur/métabolisme , Lipopolysaccharides/pharmacologie , Inhibiteurs de la cyclooxygénase 2/pharmacologie , Jonctions communicantes
10.
Cell Death Dis ; 14(9): 594, 2023 09 06.
Article de Anglais | MEDLINE | ID: mdl-37673867

RÉSUMÉ

Parkinson's disease (PD) is pathologically manifested by the aggregation of α-synuclein, which has been envisioned as a promising disease-modifying target for PD. Here, we identified 20C, a bibenzyl compound derived from Gastrodia elata, able to inhibit the aggregation of A53T variants of α-synuclein directly in vitro. Computational analysis revealed that 20C binds to cavities in mature α-synuclein fibrils, and it indeed displays a strong interaction with α-synuclein and reduced their ß-sheet structure by microscale thermophoresis and circular dichroism, respectively. Moreover, incubating neural cells with 20C reduced the amounts of α-synuclein inclusions significantly. The treatment of A53T α-Syn transgenic mice with 20C significantly reduces the toxic α-synuclein levels, improves behavioral performance, rescues dopaminergic neuron, and enhances functional connections between SNc and PD associated brain areas. The transcriptome analysis of SNc demonstrated that 20C improves mitochondrial dynamics, which protects mitochondrial morphology and function against α-synuclein induced degeneration. Overall, 20C appears to be a promising candidate for the treatment of PD.


Sujet(s)
Gastrodia , Maladie de Parkinson , Animaux , Souris , alpha-Synucléine/génétique , Maladie de Parkinson/traitement médicamenteux , Encéphale , Neurones dopaminergiques , Souris transgéniques
11.
Neurochem Int ; 169: 105584, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37454817

RÉSUMÉ

Stroke, the third leading cause of death and disability worldwide, is classified into ischemic or hemorrhagic, in which approximately 85% of strokes are ischemic. Ischemic stroke occurs as a result of arterial occlusion due to embolus or thrombus, with ischemia in the perfusion territory supplied by the occluded artery. The traditional concept that ischemic stroke is solely a vascular occlusion disorder has been expanded to include the dynamic interaction between microglia, astrocytes, neurons, vascular cells, and matrix components forming the "neurovascular unit." Acute ischemic stroke triggers a wide spectrum of neurovascular disturbances, glial activation, and secondary neuroinflammation that promotes further injury, ultimately resulting in neuronal death. Microglia, as the resident macrophages in the central nervous system, is one of the first responders to ischemic injury and plays a significant role in post-ischemic neuroinflammation. In this review, we reviewed the mechanisms of microglia in multiple stages of post-ischemic neuroinflammation development, including acute, sub-acute and chronic phases of stroke. A comprehensive understanding of the dynamic variation and the time-dependent role of microglia in post-stroke neuroinflammation could aid in the search for more effective therapeutics and diagnostic strategies for ischemic stroke.


Sujet(s)
Encéphalopathie ischémique , Accident vasculaire cérébral ischémique , Accident vasculaire cérébral , Humains , Microglie , Maladies neuro-inflammatoires , Accident vasculaire cérébral/thérapie , Macrophages
12.
Acta Pharmacol Sin ; 44(10): 1935-1947, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37198412

RÉSUMÉ

Chemokine receptor 5 (CCR5) is one of the main co-receptors of HIV-1, and has been found to be a potential therapeutic target for stroke. Maraviroc is a classic CCR5 antagonist, which is undergoing clinical trials against stroke. As maraviroc shows poor blood-brain barrier (BBB) permeability, it is of interest to find novel CCR5 antagonists suitable for neurological medication. In this study we characterized the therapeutic potential of a novel CCR5 antagonist A14 in treating ischemic stroke mice. A14 was discovered in screening millions compounds in the Chemdiv library based on the molecular docking diagram of CCR5 and maraviroc. We found that A14 dose-dependently inhibited the CCR5 activity with an IC50 value of 4.29 µM. Pharmacodynamic studies showed that A14 treatment exerted protective effects against neuronal ischemic injury both in vitro and vivo. In a SH-SY5Y cell line overexpressing CCR5, A14 (0.1, 1 µM) significantly alleviated OGD/R-induced cell injury. We found that the expression of CCR5 and its ligand CKLF1 was significantly upregulated during both acute and recovery period in focal cortical stroke mice; oral administration of A14 (20 mg·kg-1·d-1, for 1 week) produced sustained protective effect against motor impairment. A14 treatment had earlier onset time, lower onset dosage and much better BBB permeability compared to maraviroc. MRI analysis also showed that A14 treatment significantly reduced the infarction volume after 1 week of treatment. We further revealed that A14 treatment blocked the protein-protein interaction between CCR5 and CKLF1, increasing the activity of CREB signaling pathway in neurons, thereby improving axonal sprouting and synaptic density after stroke. In addition, A14 treatment remarkably inhibited the reactive proliferation of glial cells after stroke and reduced the infiltration of peripheral immune cells. These results demonstrate that A14 is a promising novel CCR5 antagonist for promoting neuronal repair after ischemic stroke. A14 blocked the protein-protein interaction between CKLF1 and CCR5 after stroke by binding with CCR5 stably, improved the infarct area and promoted motor recovery through reversing the CREB/pCREB signaling which was inhibited by activated CCR5 Gαi pathway, and benefited to the dendritic spines and axons sprouting.


Sujet(s)
Antagonistes des récepteurs CCR5 , Accident vasculaire cérébral ischémique , Neuroblastome , Accident vasculaire cérébral , Animaux , Humains , Souris , Accident vasculaire cérébral ischémique/traitement médicamenteux , Maraviroc/usage thérapeutique , Maraviroc/pharmacologie , Simulation de docking moléculaire , Récepteurs CCR5/métabolisme , Accident vasculaire cérébral/traitement médicamenteux , Antagonistes des récepteurs CCR5/composition chimique , Antagonistes des récepteurs CCR5/pharmacologie
13.
J Neuroinflammation ; 20(1): 97, 2023 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-37098609

RÉSUMÉ

Ischemic stroke is characterized by the presence of reactive microglia. However, its precise involvement in stroke etiology is still unknown. We used metabolic profiling and showed that chemokine like factor 1 (CKLF1) causes acute microglial inflammation and metabolic reprogramming from oxidative phosphorylation to glycolysis, which was reliant on the AMP-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR)-hypoxia inducible factor 1α (HIF-1α) signaling pathway. Once activated, microglia enter a chronic tolerant state as a result of widespread energy metabolism abnormalities, which reduces immunological responses, including cytokine release and phagocytosis. Metabolically dysfunctional microglia were also found in mice using genome-wide RNA sequencing after chronic administration of CKLF1, and there was a decrease in the inflammatory response. Finally, we showed that the loss of CKLF1 reversed the defective immune response of microglia, as indicated by the maintenance its phagocytosis to neutrophils, thereby mitigating the long-term outcomes of ischemic stroke. Overall, CKLF1 plays a crucial role in the relationship between microglial metabolic status and immune function in stroke, which prepares a potential therapeutic strategy for ischemic stroke.


Sujet(s)
Accident vasculaire cérébral ischémique , Accident vasculaire cérébral , Animaux , Souris , Cytokines/métabolisme , Tolérance immunitaire , Accident vasculaire cérébral ischémique/métabolisme , Mammifères/métabolisme , Microglie/métabolisme , Accident vasculaire cérébral/métabolisme
14.
J Pharm Pharmacol ; 75(5): 686-692, 2023 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-36892979

RÉSUMÉ

OBJECTIVES: Fluoxetine has been used as the first line for the therapy of depression. However, lack of therapeutic efficacy and time lag still limit the application of fluoxetine. Gap junction dysfunction is a potentially novel pathogenic mechanism for depression. To clarify the mechanism underlying these limitations, we investigated whether gap junction was related to the antidepressant effects of fluoxetine. METHODS AND KEY FINDINGS: After chronic unpredictable stress (CUS), animals showed decreases in gap junction intracellular communication (GJIC). Treatment with fluoxetine 10 mg/kg significantly improved GJIC and anhedonia of rats until six days. These results indicated that fluoxetine improved gap junction indirectly. Furthermore, to test the role of gap junction on antidepressant effects of fluoxetine, we blocked gap junction using carbenoxolone (CBX) infusion in the prefrontal cortex. CBX dampened fluoxetine-induced decrease in immobility time of mice in tail suspension test (TST). CONCLUSIONS: Our study suggested that gap junction dysfunction blocks antidepressant effects of fluoxetine, contributing to understanding the mechanism underlying the time lag of fluoxetine.


Sujet(s)
Antidépresseurs , Fluoxétine , Rats , Souris , Animaux , Fluoxétine/pharmacologie , Antidépresseurs/pharmacologie , Jonctions communicantes , Suspension des membres postérieurs , Dépression/traitement médicamenteux , Modèles animaux de maladie humaine
15.
J Pharm Pharmacol ; 75(3): 437-444, 2023 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-36734364

RÉSUMÉ

OBJECTIVES: Ginsenoside Rg1 (Rg1) has been well-documented to be effective against ischemic/reperfusion (I/R) injury. However, whether it has therapeutic effect on delayed neuronal death is still unclear. The aim of this study is to investigate the effect of Rg1 on delayed neuronal death and elucidate its underlying mechanism. METHODS: Delayed neuronal death model was prepared by global cerebral ischemia-reperfusion in rats, Rg1 was intravenously administered once a day. Nissl and Fluoro Jade B staining were carried out to evaluate the effect of Rg1 on delayed neuronal death. Western blot and qPCR were used to investigate the levels of HBXIP and Survivin. HBXIP/Survivin complex was observed by co-immunoprecipitation. AAV-CMV-shRNA (HBXIP) was used to observe the role of HBXIP on delayed neuronal death improved by Rg1. KEY FINDINGS: Rg1 attenuated delayed neuronal death at the dose of 20 mg/kg, which also improved the mRNA and protein levels of HBXIP, as well as Survivin. Moreover, administration of Rg1 promoted the formation of HBXIP/Survivin complex, which contributed to the reduction of caspases signaling pathway. Knockdown of HBXIP abolished the alleviation of DND and inhibition of caspase cascade induced by Rg1. CONCLUSIONS: Rg1 alleviated delayed neuronal death by promoting anti-apoptosis effect by HBXIP/Survivin complex.


Sujet(s)
Encéphalopathie ischémique , Ginsénosides , Lésion d'ischémie-reperfusion , Rats , Animaux , Régulation positive , Survivine , Rat Sprague-Dawley , Lésion d'ischémie-reperfusion/traitement médicamenteux , Ginsénosides/pharmacologie , Encéphalopathie ischémique/traitement médicamenteux , Reperfusion
16.
Int Immunopharmacol ; 116: 109781, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36720195

RÉSUMÉ

Chemokines, as small molecular proteins, play a crucial role in the immune and inflammatory responses after stroke. A large amount of evidence showed chemokines and their receptors were increasingly recognized as potential targets for stroke treatment, which were involved in the processing of neovascularization, neurogenesis, and neural network reconstruction. In this review, we summarized the characteristics of chemokine alterations throughout the post-stroke nerve repair phase to gain insight into the pathological mechanisms of chemokines and find effective therapeutic targets for stroke.


Sujet(s)
Récepteurs aux chimiokines , Accident vasculaire cérébral , Humains , Récepteurs aux chimiokines/métabolisme , Chimiokines/métabolisme
17.
Int Immunopharmacol ; 109: 108733, 2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-35525233

RÉSUMÉ

Microglia are the earliest activated and the longest lasting immune cells after stroke, and they participate in almost all the pathological reactions after stroke. However, their regulatory mechanism has not been fully elucidated. Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor that is mainly expressed in microglia of the central nervous system. The receptor plays an important role in regulating microglia energy metabolism and phenotypic transformation. At present, TREM2 has been developed as a potential target for AD, coronary atherosclerosis and other diseases. However, TREM2 does not provide a systematic summary of the functional transformation and intrinsic molecular mechanisms of microglia after stroke. In this paper, we have summarized the functional changes of TREM2 in microglia after stroke in recent years, and found that TREM2 has important effects on energy metabolism, phagocytosis and anti-inflammatory function of microglia after stroke, suggesting that TREM2 is a potential therapeutic target for the treatment of stroke.


Sujet(s)
Accident vasculaire cérébral ischémique , Glycoprotéines membranaires , Récepteurs immunologiques , Humains , Accident vasculaire cérébral ischémique/métabolisme , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Microglie/métabolisme , Cellules myéloïdes , Phagocytose , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme
18.
Acta Pharmacol Sin ; 43(4): 797-810, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-34226665

RÉSUMÉ

Newborns suffering from hypoxia-ischemia (HI) brain injury still lack effective treatment. Proline-rich tyrosine kinase 2 (Pyk2) is a non-receptor tyrosine kinase, which is highly correlated with transient ischemic brain injury in adult. In this study, we investigated the role of Pyk2 in neonatal HI brain injury. HI was induced in postnatal day 7 mouse pups by unilateral common carotid artery ligation followed by hypoxic exposure. Pyk2 interference lentivirus (LV-Pyk2 shRNA) was constructed and injected into unilateral cerebral ventricle of neonatal mice before HI. Infarct volume, pathological changes, and neurological behaviors were assessed on postnatal day 8-14. We showed that the phosphorylation level of Pyk2 was significantly increased in neonatal brain after HI, whereas LV-Pyk2 shRNA injection significantly attenuated acute HI brain damage and improved neurobehavioral outcomes. In oxygen-glucose deprivation-treated cultured cortical neurons, Pyk2 inhibition significantly alleviated NMDA receptor-mediated excitotoxicity; similar results were also observed in neonatal HI brain injury. We demonstrated that Pyk2 inhibition contributes to the long-term cerebrovascular recovery assessed by laser speckle contrast imaging, but cognitive function was not obviously improved as evaluated in Morris water maze and novel object recognition tests. Thus, we constructed lentiviral LV-HIF-Pyk2 shRNA, through which HIF-1α promoter-mediated interference of Pyk2 would occur during the anoxic environment. Intracerebroventricular injection of LV-HIF-Pyk2 shRNA significantly improved long-term recovery of cognitive function in HI-treated neonatal mice. In conclusion, this study demonstrates that Pyk2 interference protects neonatal brain from hypoxic-ischemic injury. HIF-1α promoter-mediated hypoxia conditional control is a useful tool to distinguish between hypoxic period and normal period. Pyk2 is a promising drug target for potential treatment of neonatal HI brain injury.


Sujet(s)
Lésions encéphaliques , Hypoxie-ischémie du cerveau , Animaux , Animaux nouveau-nés , Encéphale/anatomopathologie , Lésions encéphaliques/anatomopathologie , Focal adhesion kinase 2/pharmacologie , Hypoxie-ischémie du cerveau/traitement médicamenteux , Souris
19.
Acta Pharmacol Sin ; 43(5): 1217-1230, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-34385606

RÉSUMÉ

The phenotypic transformation of microglia in the ischemic penumbra determines the outcomes of ischemic stroke. Our previous study has shown that chemokine-like-factor 1 (CKLF1) promotes M1-type polarization of microglia. In this study, we investigated the cellular source and transcriptional regulation of CKLF1, as well as the biological function of CKLF1 in ischemic penumbra of rat brain. We showed that CKLF1 was significantly up-regulated in cultured rat cortical neurons subjected to oxygen-glucose deprivation/reoxygenation (ODG/R) injury, but not in cultured rat microglia, astrocytes and oligodendrocytes. In a rat model of middle cerebral artery occlusion, we found that CKLF1 was up-regulated and co-localized with neurons in ischemic penumbra. Furthermore, the up-regulated CKLF1 was accompanied by the enhanced nuclear accumulation of NF-κB. The transcriptional activity of CKLF1 was improved by overexpression of NF-κB in HEK293T cells, whereas application of NF-κB inhibitor Bay 11-7082 (1 µM) abolished it, caused by OGD/R. By using chromatin-immunoprecipitation (ChIP) assay we demonstrated that NF-κB directly bound to the promoter of CKLF1 (at a binding site located at -249 bp to -239 bp of CKLF1 promoter region), and regulated the transcription of human CKLF1. Moreover, neuronal conditional medium collected after OGD/R injury or CKLF1-C27 (a peptide obtained from secreted CKLF1) induced the M1-type polarization of microglia, whereas the CKLF1-neutralizing antibody (αCKLF1) or NF-κB inhibitor Bay 11-7082 abolished the M1-type polarization of microglia. Specific knockout of neuronal CKLF1 in ischemic penumbra attenuated neuronal impairments and M1-type polarization of microglia caused by ischemic/reperfusion injury, evidenced by inhibited levels of M1 marker CD16/32 and increased expression of M2 marker CD206. Application of CKLF1-C27 (200 nM) promoted the phosphorylation of p38 and JNK in microglia, whereas specific depletion of neuronal CKLF1 in ischemic penumbra abolished ischemic/reperfusion-induced p38 and JNK phosphorylation. In summary, CKLF1 up-regulation in neurons regulated by NF-κB is one of the crucial mechanisms to promote M1-type polarization of microglia in ischemic penumbra.


Sujet(s)
Encéphalopathie ischémique , Accident vasculaire cérébral , Animaux , Encéphale/métabolisme , Encéphalopathie ischémique/métabolisme , Chimiokines/métabolisme , Cellules HEK293 , Humains , Protéines à domaine MARVEL , Microglie/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Neurones/métabolisme , Rats , Accident vasculaire cérébral/métabolisme , Régulation positive
20.
Acta Pharmacol Sin ; 43(3): 563-576, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-34103690

RÉSUMÉ

Myelin damage and abnormal remyelination processes lead to central nervous system dysfunction. Glial activation-induced microenvironment changes are characteristic features of the diseases with myelin abnormalities. We previously showed that ginsenoside Rg1, a main component of ginseng, ameliorated MPTP-mediated myelin damage in mice, but the underlying mechanisms are unclear. In this study we investigated the effects of Rg1 and mechanisms in cuprizone (CPZ)-induced demyelination mouse model. Mice were treated with CPZ solution (300 mg· kg-1· d-1, ig) for 5 weeks; from week 2, the mice received Rg1 (5, 10, and 20 mg· kg-1· d-1, ig) for 4 weeks. We showed that Rg1 administration dose-dependently alleviated bradykinesia and improved CPZ-disrupted motor coordination ability in CPZ-treated mice. Furthermore, Rg1 administration significantly decreased demyelination and axonal injury in pathological assays. We further revealed that the neuroprotective effects of Rg1 were associated with inhibiting CXCL10-mediated modulation of glial response, which was mediated by NF-κB nuclear translocation and CXCL10 promoter activation. In microglial cell line BV-2, we demonstrated that the effects of Rg1 on pro-inflammatory and migratory phenotypes of microglia were related to CXCL10, while Rg1-induced phagocytosis of microglia was not directly related to CXCL10. In CPZ-induced demyelination mouse model, injection of AAV-CXCL10 shRNA into mouse lateral ventricles 3 weeks prior CPZ treatment occluded the beneficial effects of Rg1 administration in behavioral and pathological assays. In conclusion, CXCL10 mediates the protective role of Rg1 in CPZ-induced demyelination mouse model. This study provides new insight into potential disease-modifying therapies for myelin abnormalities.


Sujet(s)
Chimiokine CXCL10/antagonistes et inhibiteurs , Maladies démyélinisantes/anatomopathologie , Ginsénosides/pharmacologie , Animaux , Cuprizone/pharmacologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Hypocinésie/anatomopathologie , Médiateurs de l'inflammation/métabolisme , Mâle , Souris , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Panax/composition chimique , Panax/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Petit ARN interférent/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE