Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Pharmacother ; 176: 116778, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38788601

ABSTRACT

The incidence of cerebral infarction triggered by abnormal glucose tolerance has increased; however, the relationship between glucose concentration in the brain and the detailed mechanism of post ischemic cell death remains unclear. Nicotinamide phosphoribosyltransferase (NAMPT), an adipocytokine, is the rate-limiting enzyme for NAD+ synthesis in the salvage pathway. Although NAMPT activation prevents neuronal injury, the relationship between NAMPT activity, glucose metabolism disorders, and cerebral ischemia-induced neuronal cell death is unknown. In this study, we determined changes in NAMPT on cerebral ischemic injuries with diabetes using a db/db mouse model of type 2 diabetes and then identified the underlying mechanisms using Neuro2a cells. The expression of inflammatory cytokine mRNAs was increased in db/db and db/+ middle cerebral artery occlusion and reperfusion (MCAO/R) mice. Although NeuN-positive cells were decreased after MCAO/R, the number of NAMPT and NeuN double-positive cells in NeuN-positive neuronal cells increased in db/db MCAO/R mice. Next, the role of NAMPT in Neuro2a cells under conditions of high glucose (HGC) and oxygen-glucose deprivation (OGD), which mimics diabetes-complicated cerebral infarction, was examined. Treatment with P7C3-A20, a NAMPT activator, suppressed the decrease in cell viability caused by HGC/OGD; however, there were no significant differences in the levels of cleaved caspase-3 and Bax proteins. Moreover, increased FoxO3a and LC3-II levels after HGC/OGD were inhibited by P7C3-A20 treatment. Our findings indicate that NAMPT activation is associated with neuronal survival under ischemic conditions with abnormal glucose tolerance through the regulation of FoxO3a/LC3.

2.
Exp Neurol ; 371: 114584, 2024 01.
Article in English | MEDLINE | ID: mdl-37884188

ABSTRACT

Ischemic stroke in patients with abnormal glucose tolerance results in poor outcomes. Nicotinamide phosphoribosyltransferase (NAMPT), an adipocytokine, exerts neuroprotective effects. However, the pathophysiological role of NAMPT after ischemic stroke with diabetes and the relationship of NAMPT with cerebrovascular lesions are unclear. The purpose of this study was to clarify the pathophysiological role of NAMPT in cerebral ischemia with diabetes, using db/db mice as a type 2 diabetes animal model. The number of degenerating neurons increased after middle cerebral artery occlusion and reperfusion (MCAO/R) in db/db mice compared with the degenerating neurons in db/+ mice. Extracellular NAMPT (eNAMPT) levels, especially monomeric eNAMPT, increased significantly in db/db MCAO/R mice but not db/+ mice in isolated brain microvessels. The increased eNAMPT levels were associated with increased expression of inflammatory cytokine mRNA. Immunohistochemical analysis demonstrated that NAMPT colocalized with GFAP-positive cells after MCAO/R. In addition, both dimeric and monomeric eNAMPT levels increased in the conditioned medium of primary cortical astrocytes under high glucose conditions subsequent oxygen/glucose deprivation. Our findings are the first to demonstrate the ability of increased monomeric eNAMPT to induce inflammatory responses in brain microvessels, which may be located near astrocyte foot processes.


Subject(s)
Diabetes Mellitus, Type 2 , Ischemic Stroke , Stroke , Animals , Humans , Mice , Cytokines , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Infarction, Middle Cerebral Artery/complications , Nicotinamide Phosphoribosyltransferase/metabolism , Stroke/complications , Stroke/pathology
3.
Biomed Pharmacother ; 168: 115683, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37832402

ABSTRACT

Following brain injury, neural stem cells (NSCs) can generate mature neurons and replace damaged cells. However, the capacity of endogenous NSCs to self-repair from injured brain is limited as most NSCs die before becoming mature neurons. Therefore, a boosting endogenous NSCs by pharmacological support offers the potential to repair the damaged brain. Recently, small molecules have hold considerable promise for neuron regeneration and repair as they can penetrate the blood-brain barrier easily. Senkyunolide I (SEI) is a bioactive constituent derived from traditional Chinese medicines Ligusticum chuanxiong Hort. and Angelica sinensis (Oliv.) Diels, and was found to able to prevent ischemic stroke. This study examined the effects of SEI on the proliferation and neuronal lineage differentiation of prepared neural stem/progenitor cells (NS/PCs). The NS/PC proliferation was determined by 2-(4-iodophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt, and neurosphere formation assays. The NS/PC differentiation was also investigated by immunocytochemistry, and western blotting was employed to measure phosphorylated Akt (pAkt) and GSK-3ß (pGSK-3ß), and active-ß-catenin protein levels. We showed that the NS/PC proliferation was enhanced after SEI exposure. Elevated cell numbers were also observed in neurospheres, which were incubated with SEI for 3 days, whereas the NS/PC differentiation was decreased after SEI exposure for 5 days. Furthermore, SEI upregulated pAkt/Akt and active-ß-catenin levels and increased NS/PC proliferation after SEI treatment was reversed by phosphatidylinositol 3-kinase inhibitor LY294002. downregulated differentiated processes. Thus, SEI promoted the NS/PC proliferation and suppressed NS/PC differentiation into neurons and/or astrocytes, therefore SEI could be an interesting and promising candidate for stimulating NSCs.


Subject(s)
Neural Stem Cells , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Cell Proliferation , Neural Stem Cells/metabolism , Cell Differentiation
4.
Int J Mol Sci ; 23(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35216064

ABSTRACT

We previously demonstrated that sivelestat, a selective neutrophil elastase inhibitor, attenuates the cleavage of progranulin (PGRN) and ischemia-induced cell injury in the brain. To obtain further insight into the role of PGRN, in the present study we evaluated the direct effects of sivelestat and recombinant PGRN (rPGRN) on the proliferation and differentiation of neural stem cells in cultures of neural stem/progenitor cells (NS/PC) under the ischemic condition in vitro. We demonstrated that oxygen/glucose deprivation (OGD)-induced cell proliferation of NS/PC was increased by rPGRN treatment. In addition, this increase was accompanied by increased phosphorylation of Akt and GSK-3ß (Ser9) after OGD. But none of these responses occurred by treatment with sivelestat. Therefore, activation of the Akt/GSK-3ß pathway could well be involved in this proliferative effect of rPGRN. Although OGD and reoxygenation-induced changes in the differentiation of NS/PC into neurons or astrocytes was not affected by treatment with rPGRN or sivelestat, it is noteworthy that rPGRN enhanced neurite outgrowth of ß3-tubulin-positive neurons that had differentiated from the NS/PC. These findings suggest that enhancement of proliferation of endogenous NS/PC and neurite outgrowth of differentiated neurons from NS/PC by PGRN could be useful for a new therapeutic approach for cerebral ischemia.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Glucose/metabolism , Neural Stem Cells/drug effects , Oxygen/metabolism , Progranulins/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Male , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar
5.
Front Oncol ; 11: 711220, 2021.
Article in English | MEDLINE | ID: mdl-34513690

ABSTRACT

Development of new therapeutic strategies for breast cancer is urgently needed due to the sustained emergence of drug resistance, tumor recurrence and metastasis. To gain a novel insight into therapeutic approaches to fight against breast cancer, the cytocidal effects of hellebrigenin (Helle) and arenobufagin (Areno) were investigated in human estrogen receptor (ER)-positive breast cancer cell line MCF-7 and triple-negative breast cancer cell line MDA-MB-231. Helle exhibited more potent cytotoxicity than Areno in both cancer cells, and MCF-7 cells were more susceptible to both drugs in comparison with MDA-MB-231 cells. Apoptotic-like morphological characteristics, along with the downregulation of the expression level of Bcl-2 and Bcl-xL and the upregulation of the expression level of Bad, were observed in Helle-treated MCF-7 cells. Helle also caused the activation of caspase-8, caspase-9, along with the cleavage of poly(ADP-ribose) polymerase in MCF-7 cells. Helle-mediated necrosis-like phenotype, as evidenced by the increased propidium iodide (PI)-positive cells was further observed. G2/M cell cycle arrest was also induced by Helle in the cells. Upregulation of the expression level of p21 and downregulation of the expression level of cyclin D1, cyclin E1, cdc25C and survivin were observed in MCF-7 cells treated with Helle and occurred in parallel with G2/M arrest. Autophagy was triggered in MCF-7 cells and the addition of wortmannin or 3-MA, two well-known autophagy inhibitors, slightly but significantly rescued the cells. Furthermore, similar alterations of some key molecules associated with the aforementioned biological phenomena were observed in MDA-MB-231 cells. Intriguingly, the numbers of PI-positive cells in Helle-treated MCF-7 cells were significantly reduced by wortmannin and 3-MA, respectively. In addition, Helle-triggered G2/M arrest was significantly corrected by wortmannin, suggesting autophagy induction contributed to Helle-induced cytotoxicity of breast cancer cells by modulating necrosis and cell cycle arrest. Collectively, our results suggested potential usefulness of both Helle and Areno in developing therapeutic strategies to treat patients with different types of breast cancer, especially ER-positive breast cancer.

6.
Biomed Pharmacother ; 140: 111696, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34044281

ABSTRACT

Glutamate-induced neurotoxicity is one of the most important pathogenic mechanisms in neurological diseases and is widely used as an in vitro model for ischemic stroke. Senkyunolide I (SEI), an active constituent derived from traditional Chinese medicine Ligusticum chuanxiong Hort. and Angelica sinensis (Oliv.) Diels, has been shown to have beneficial effects against focal cerebral ischemia-reperfusion in rats. However, the mechanisms underlying SEI-mediated neuroprotection remain not well understood. Thus, we explored the influence of SEI in glutamate-mediated injury to mouse neuroblastoma (Neuro2a) cells and determined the mechanisms involved. Neuro2a cells were treated with SEI under exposure to glutamate for 24 h. Cell viability was assessed by using WST-1 reagents, and apoptosis was evaluated using Annexin V-FITC and a PI double staining kit. The protein expression levels of p-AKT, AKT, p-GSK3ß, GSK3ß, p-p38, p38, p-ERK, ERK, p-JNK, JNK, Bcl-2, Bax, Bcl-xl, p-Bad, Bad, p53, and cleaved caspase-3 were determined by Western blot analysis. Glutamate significantly decreased cell viability and elevated the level of apoptosis. Treatment with SEI reversed those effects. Furthermore, the expression of p-JNK/JNK and cleaved caspase-3 were also reduced after treatment with SEI. Our findings demonstrate that SEI protected Neuro2a cells against glutamate toxicity by regulating JNK/caspase-3 pathway and apoptosis. Thus, SEI maybe a promising candidate for neuroprotection.


Subject(s)
Apoptosis/drug effects , Benzofurans/pharmacology , Caspase 3/metabolism , Cell Death/drug effects , Glutamic Acid/metabolism , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cell Survival/drug effects , Cells, Cultured , Mice , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroprotection/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction/drug effects
7.
Mol Cell Neurosci ; 113: 103625, 2021 06.
Article in English | MEDLINE | ID: mdl-33933589

ABSTRACT

In a previous study, we demonstrated that neutrophil elastase is activated in the brain parenchyma after cerebral ischemia, which enzyme cleaves progranulin (PGRN), an anti-inflammatory factor. In that study, we also found that sivelestat, a selective neutrophil elastase inhibitor, attenuates ischemia-induced inflammatory responses. However, it was not clear whether this anti-inflammatory effect was due to the direct effect of sivelestat. In this study, we evaluated the effects of sivelestat or recombinant PGRN (rPGRN) on cell injuries in cultured neurons, astrocytes, and microglia under oxygen/glucose deprivation (OGD) conditions. We demonstrated that OGD-induced neuronal cell injury, astrocyte activation, and increased proinflammatory cytokines caused by microglial activation, were suppressed by rPGRN treatment, whereas sivelestat had no effect on any of these events. These results indicate that the anti-inflammatory responses after in vivo cerebral ischemia were not due to the direct action of sivelestat but due to the suppression of PGRN cleavage by inhibition of elastase activity. It was also suggested that the pleiotropic effect of rPGRN could be attributed to the differentiation of M1 microglia into anti-inflammatory type M2 microglia. Therefore, the inhibition of PGRN cleavage by sivelestat could contribute to the establishment of a new therapeutic approach for cerebral ischemia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Ischemia/metabolism , Glycine/analogs & derivatives , Neuroglia/drug effects , Neurons/drug effects , Progranulins/pharmacology , Serine Proteinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Animals , Cell Hypoxia , Cells, Cultured , Cytokines/metabolism , Glycine/pharmacology , Male , Neuroglia/metabolism , Neurons/metabolism , Pancreatic Elastase/antagonists & inhibitors , Rats , Rats, Wistar , Recombinant Proteins/pharmacology
8.
Sci Rep ; 9(1): 11782, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409872

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor has been implicated in several neurodegenerative diseases, including stroke. Low-density lipoprotein receptor-related protein 1 (LRP1) plays pivotal roles in endocytosis and signaling in the cell. Immature LRP1 is processed by furin in the trans-Golgi network (TGN) and transported to the cell surface as its mature form. Activation of mature LRP1 exerts a protective effect against glutamate-induced degeneration of the rat retinal ganglion cells, as was shown in our previous study. However, the roles of LRP1 in the pathogenesis of excitotoxic neuronal injuries remain to be determined. The aim of this present study was to achieve further insight into the pathophysiologic roles of LRP1 after excitotoxic neuronal injuries. Our findings are the first to demonstrate that LRP1 was significantly cleaved by furin after cerebral ischemia in rats as well as after exposure of cultured cortical neurons to NMDA. It was noteworthy that the intracellular domain (ICD) of LRP1 was co-localized with TGN and furin. Furthermore, a furin inhibitor inhibited the cleavage of LRP1 and co-localization of LRP1-ICD with TGN or furin. Our findings suggest that furin-mediated cleavage of LRP1 and changes in the localization of LRP1-ICD were involved in the excitotoxic neuronal injury.


Subject(s)
Brain Ischemia/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , N-Methylaspartate/metabolism , Stroke/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Endocytosis/drug effects , Furin/metabolism , Humans , N-Methylaspartate/pharmacology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Rats , Receptors, LDL/genetics , Signal Transduction/drug effects , Stroke/metabolism , Stroke/pathology , trans-Golgi Network/drug effects , trans-Golgi Network/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...