Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38411782

ABSTRACT

Cerebral ischemic preconditioning (CIP) has been shown to improve brain ischemic tolerance against subsequent lethal ischemia. Reactive astrocytes play important roles in cerebral ischemia-reperfusion. Recent studies have shown that reactive astrocytes can be polarized into neurotoxic A1 phenotype (C3d) and neuroprotective A2 phenotype (S100A10). However, their role in CIP remains unclear. Here, we focused on the role of N-myc downstream-regulated gene 2 (NDRG2) in regulating the transformation of A1/A2 astrocytes and promoting to brain ischemic tolerance induced by CIP. A Sprague Dawley rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) was used. Rats were divided into the following six groups: (1) sham group; (2) CIP group: left middle cerebral artery was blocked for 10 min; (3) MCAO/R group: left middle cerebral artery was blocked for 90 min; (4) CIP + MCAO/R group: CIP was performed 72 h before MCAO/R; (5) AAV-NDRG2 + CIP + MCAO/R group: adeno-associated virus (AAV) carrying NDRG2 was administered 14 days before CIP + MCAO/R; (6) AAV-Ctrl + CIP + MCAO/R group: empty control group. The rats were subjected to neurological evaluation 24 h after the above treatments, and then were sacrificed for 2, 3, 5-triphenyltetraolium chloride staining, thionin staining, immunofluorescence and western blot analysis. In CIP + MCAO/R group, the neurological deficit scores decreased, infarct volume reduced, and neuronal density increased compared with MCAO/R group. Notably, CIP significantly increased S100A10 expression and the number of S100A10+/GFAP+ cells, and also increased NDRG2 expression. MCAO/R significantly decreased S100A10 expression and the number of S100A10+/GFAP+ cells yet increased C3d expression and the number of C3d+/GFAP+ cells and NDRG2 expression, and these trends were reversed by CIP + MCAO/R. Furthermore, over-expression of NDRG2 before CIP + MCAO/R, the C3d expression and the number of C3d+/GFAP+ cells increased, while S100A10 expression and the number of S100A10+/GFAP+ cells decreased. Meanwhile, over-expression of NDRG2 blocked the CIP-induced brain ischemic tolerance. Taken together, these results suggest that CIP exerts neuroprotective effects against ischemic injury by suppressing A1 astrocyte polarization and promoting A2 astrocyte polarization via inhibiting NDRG2 expression.

2.
Mol Neurobiol ; 61(4): 2336-2356, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37875707

ABSTRACT

Our previous study has proved that the Klotho up-regulation participated in cerebral ischemic preconditioning (CIP)-induced brain ischemic tolerance. However, the exact neuroprotective mechanism of Klotho in CIP remains unclear. We explored the hypothesis that STAT4-mediated Klotho up-regulation contributes to the CIP-induced brain ischemic tolerance via inhibiting neuronal pyroptosis. Firstly, the expressions of pyroptosis-associated proteins (i.e., NLRP3, GSDMD, pro-caspase-1, and cleaved caspase-1) in hippocampal CA1 region were determined during the process of brain ischemic tolerance. We found the expression of pyroptosis-associated proteins was significantly up-regulated in the ischemic insult (II) group, and showed no significant changes in the CIP group. The expression level of each pyroptosis-associated proteins was lower in the CIP + II group than that in the II group. Inhibition of Klotho expression increased the expression of pyroptosis-associated proteins in the CIP + II group and blocked the CIP-induced brain ischemic tolerance. Injection of Klotho protein decreased the expression of pyroptosis-associated proteins in the II group, and protected neurons from ischemic injury. Secondly, the transcription factor STAT4 of Klotho was identified by bioinformatic analysis. Double luciferase reporter gene assay and chromatin immunoprecipitation assay showed STAT4 can bind to the site between nt - 881 and - 868 on the Klotho promoter region and positively regulates Klotho expression. Moreover, we found CIP significantly enhanced the expression of STAT4. Knockdown STAT4 suppressed Klotho up-regulation after CIP and blocked the CIP-induced brain ischemic tolerance. Collectively, it can be concluded that STAT4-mediated the up-regulation of Klotho contributed to the brain ischemic tolerance induced by CIP via inhibiting pyroptosis.


Subject(s)
Brain Ischemia , Ischemic Preconditioning , Rats , Animals , Rats, Wistar , Up-Regulation , Pyroptosis , STAT4 Transcription Factor/metabolism , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Neurons/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
3.
Mol Neurobiol ; 61(4): 2270-2282, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37870679

ABSTRACT

The morbidity rate of ischemic stroke is increasing annually with the growing aging population in China. Astrocytes are ubiquitous glial cells in the brain and play a crucial role in supporting neuronal function and metabolism. Increasing evidence shows that the impairment or loss of astrocytes contributes to neuronal dysfunction during cerebral ischemic injury. The mitochondrion is increasingly recognized as a key player in regulating astrocyte function. Changes in astrocytic mitochondrial function appear to be closely linked to the homeostasis imbalance defects in glutamate metabolism, Ca2+ regulation, fatty acid metabolism, reactive oxygen species, inflammation, and copper regulation. Here, we discuss the role of astrocytic mitochondria in the pathogenesis of brain ischemic injury and their potential as a therapeutic target.


Subject(s)
Brain Injuries , Brain Ischemia , Humans , Aged , Astrocytes/metabolism , Brain Ischemia/pathology , Brain/metabolism , Brain Injuries/metabolism , Mitochondria/metabolism
4.
Cell Mol Neurobiol ; 43(3): 1355-1367, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35900650

ABSTRACT

Cerebral ischemic preconditioning (CIP)-induced brain ischemic tolerance protects neurons from subsequent lethal ischemic insult. However, the specific mechanisms underlying CIP remain unclear. In the present study, we explored the hypothesis that peroxisome proliferator-activated receptor gamma (PPARγ) participates in the upregulation of Klotho during the induction of brain ischemic tolerance by CIP. First we investigated the expression of Klotho during the brain ischemic tolerance induced by CIP. Lethal ischemia significantly decreased Klotho expression from 6 h to 7 days, while CIP significantly increased Klotho expression from 12 h to 7 days in the hippocampal CA1 region. Inhibition of Klotho expression by its shRNA blocked the neuroprotection induced by CIP. These results indicate that Klotho participates in brain ischemic tolerance by CIP. Furthermore, we tested the role of PPARγ in regulating Klotho expression after CIP. CIP caused PPARγ protein translocation to the nucleus in neurons in the CA1 region of the hippocampus. Pretreatment with GW9962, a PPARγ inhibitor, significantly attenuated the upregulation of Klotho protein and blocked the brain ischemic tolerance induced by CIP. Taken together, it can be concluded that Klotho upregulation via PPARγ contributes to the induction of brain ischemic tolerance by CIP.


Subject(s)
Brain Ischemia , Ischemic Preconditioning , Animals , Rats , Brain Ischemia/metabolism , CA1 Region, Hippocampal , Ischemia , PPAR gamma/metabolism , Rats, Wistar , Up-Regulation
5.
DNA Cell Biol ; 41(9): 838-849, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35944278

ABSTRACT

Several studies indicated that autophagy activation participates in brain ischemic tolerance (BIT) induced by cerebral ischemic preconditioning (CIP). However, the mechanism of autophagy activation during the process still remains unclear. The present study aimed to evaluate the role of p38 MAPK-peroxisome proliferator-activated receptor γ (PPARγ) signaling cascade in autophagy during the CIP-induced BIT. The results shown that, initially, autophagy activation was observed after CIP in the model of global cerebral ischemia in rats, as was indicated by the upregulation of Beclin 1 expression, an increase in LC3-II/LC3-I ratio, the enhanced LC3 immunofluorescence, and a rise in the number of autophagosomes in the neurons of the hippocampal CA1 area. Besides, the inhibitor of autophagy 3-methyladenine obliterated the neuroprotection induced by CIP. Furthermore, the upregulation of p-p38 MAPK and PPARγ expressions was earlier than autophagy activation after CIP. In addition, pretreatment with SB203580 (the inhibitor of p38 MAPK) reversed CIP-induced PPARγ upregulation, autophagy activation, and neuroprotection. Pretreatment with GW9662 (the inhibitor of PPARγ) reversed autophagy activation and neuroprotection, while it had no effect on p-p38 MAPK upregulation induced by CIP. These data suggested that the p38 MAPK-PPARγ signaling pathway participates in autophagy activation during the induction of BIT by CIP.


Subject(s)
Brain Ischemia , Ischemic Preconditioning , Animals , Autophagy , Brain/metabolism , Brain Ischemia/metabolism , Ischemic Preconditioning/methods , PPAR gamma/genetics , PPAR gamma/metabolism , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Brain Res Bull ; 175: 224-233, 2021 10.
Article in English | MEDLINE | ID: mdl-34343641

ABSTRACT

Our previous finding suggests that p38 MAPK contributes to the GLT-1 upregulation during induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP), however, the underlying mechanism is still unclear. Here, we investigated the molecular mechanisms underlying the CIP-induced GLT-1 upregulation by using Western blotting, Co-immunoprecipitation (Co-IP), electrophoretic mobility shift assay (EMSA) and thionin staining in rat hippocampus CA1 subset. We found that application of BAY11-7082 (an inhibitor of NF-κB), or dihydrokainate (an inhibitor of GLT-1), or SB203580 (an inhibitor of p38 MAPK) could attenuate the CIP-induced neuronal protection in hippocampus CA1 region of rats. Moreover, CIP caused rapid activation of NF-κB, as evidenced by nuclear translocation of NF-κB p50 protein, which led to active p50/p65 dimer formation and increased DNA binding activity. GLT-1 was also increased after CIP. Pretreatment with BAY11-7082 blocked the CIP-induced GLT-1 upregulation. The above results suggest that NF-κB participates in GLT-1 up-regulation during the induction of brain ischemic tolerance by CIP. We also found that pretreatment with SB203580 caused significant reduction of NF-κB p50 protein in nucleus, NF-κB p50/p65 dimer nuclear translocation and DNA binding activity of NF-κB. Together, we conclude that p38 MAPK/NF-κB pathway participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance induced by CIP.


Subject(s)
Brain Ischemia/genetics , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Ischemic Preconditioning , MAP Kinase Signaling System/genetics , NF-kappa B/genetics , Animals , CA1 Region, Hippocampal/pathology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Imidazoles/pharmacology , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , MAP Kinase Signaling System/drug effects , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Neuroprotection , Nitriles/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Sulfones/pharmacology , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases
7.
J Neurochem ; 151(5): 608-625, 2019 12.
Article in English | MEDLINE | ID: mdl-31314916

ABSTRACT

Glial glutamate transporter 1 (GLT-1) plays a vital role in the induction of brain ischemic tolerance (BIT) by ischemic preconditioning (IPC). However, the mechanism still needs to be further explained. The aim of this study was to investigate whether peroxisome proliferator-activated receptor gamma (PPARγ) participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Initially, cerebral IPC induced BIT and enhanced PPARγ and GLT-1 expression in the CA1 hippocampus in rats. The ratio of nuclear/cytoplasmic PPARγ was also increased. At the same time, the up-regulation of PPARγ expression in astrocytes in the CA1 hippocampus was revealed by double immunofluorescence for PPARγ and glial fibrillary acidic protein. Then, the mechanism by which PPARγ regulates GLT-1 was studied in rat cortical astrocyte-neuron cocultures. We found that IPC [45 min of oxygen glucose deprivation (OGD)] protected neuronal survival after lethal OGD (4 h of OGD), which usually leads to neuronal death. The activation of PPARγ occurred earlier than the up-regulation of GLT-1 in astrocytes after IPC, as determined by western blot and immunofluorescence. Moreover, the preadministration of the PPARγ antagonist T0070907 or PPARγ siRNA significantly attenuated GLT-1 up-regulation and the neuroprotective effects induced by IPC in vitro. Finally, the effect of the PPARγ antagonist on GLT-1 expression and BIT was verified in vivo. We observed that the preadministration of T0070907 by intracerebroventricular injection dose-dependently attenuated the up-regulation of GLT-1 and BIT induced by cerebral IPC in rats. In conclusion, PPARγ participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Cover Image for this issue: doi: 10.1111/jnc.14532. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Brain/blood supply , Brain/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Ischemic Preconditioning , PPAR gamma/metabolism , Animals , Brain Ischemia/metabolism , In Vitro Techniques , Male , Neuroglia/metabolism , Rats , Rats, Wistar
8.
Brain Res Bull ; 147: 1-13, 2019 04.
Article in English | MEDLINE | ID: mdl-30731111

ABSTRACT

The previous studies have shown that glial glutamate transporter-1 (GLT-1) participates in cerebral ischemic injury in rats. However, the mechanism involved remains to be elucidated. This study was undertaken to investigate whether p38 MAPK was involved in regulating GLT-1 in the process. At first, it was observed that global brain ischemia for 8 min led to obvious delayed neuronal death, GLT-1 down-regulation and p-p38 MAPK up-regulation in CA1 hippocampus in rats. Then, whether p-p38 MAPK was involved in regulating GLT-1 during cerebral ischemic injury was studied in vitro. Astrocyte-neuron co-cultures exposed to oxygen and glucose deprivation (OGD) were used to mimic brain ischemia. It was observed that lethal OGD (4-h OGD) decreased GLT-1 expression and increased p-p38 MAPK expression in astrocytes. The p-p38 MAPK protein rised from 0 min to 48 h that is the end time of the observation, and the peak value was at 12 h, which was 12.45 times of the control group. Moreover, pre-administration of p38 MAPK inhibitor SB203580 or its siRNA dose-dependently increased GLT-1 expression, and meanwhile alleviated the neuronal death induced by lethal OGD. The above results indicated that p38 MAPK signaling pathway participated in regulating GLT-1 during OGD injury in vitro. Finally, back to in vivo experiment, it was found that pre-administration of SB203580 by intracerebroventricular injection dose-dependently reversed the down-regulation of GLT-1 expression and attenuated the delayed neuronal death normally induced by global brain ischemia in CA1 hippocampus in rats. Taken together, it can be concluded that the mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK.


Subject(s)
Brain Ischemia/metabolism , Excitatory Amino Acid Transporter 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Astrocytes/metabolism , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/metabolism , Cell Death , Coculture Techniques , Excitatory Amino Acid Transporter 2/physiology , Glucose/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Imidazoles/pharmacology , MAP Kinase Signaling System , Male , Neurons/metabolism , Oxygen/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
9.
Neurochem Res ; 43(10): 2016, 2018 10.
Article in English | MEDLINE | ID: mdl-30171421

ABSTRACT

The order of corresponding author was inadvertently published. Hence, the first and the second corresponding authors should be Min Zhang (hebmuzhangmin@163.com) and Jing-Ge Zhang (zhangjg001@163.com).

10.
Front Mol Neurosci ; 11: 281, 2018.
Article in English | MEDLINE | ID: mdl-30158854

ABSTRACT

Sulbactam is an atypical ß-lactam medication and reported to be neuroprotective by up-regulating glial glutamate transporter-1 (GLT-1) in rats. The present study was undertaken to study the role of p38 MAPK signal pathway in sulbactam induced up-regulation of GLT-1 expression in astrocytes and anti-ischemic effect. Neuron-astrocyte co-cultures and astrocyte cultures from neonatal Wistar rats were used. Cerebral ischemia was mimicked by oxygen-glucose deprivation (OGD). Hoechst (HO)/propidium iodide (PI) double fluorescence staining and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay were used to evaluate neuronal death and cell viability, respectively. Immunocytochemistry and Western blot were used to detect protein expressions. Sulbactam pre-incubation significantly and dose-dependently prevented neuronal death and decline in cell viability induced by OGD in neuron-astrocyte co-cultures, and upregulated GLT-1 expression in astrocyte cultures endured OGD, which suggested that sulbactam might protect neurons against OGD by up-regulating astrocytic GLT-1 expression. It was further shown that the phosphorylated-p38 MAPK expression in astrocytes was up-regulated after the sulbactam pre-incubation and this up-regulation was moderate in amplitude. Especially, the time course of the up-regulation of phosphorylated-p38 MAPK was obviously earlier than that of GLT-1, which suggested possibility that p38 MAPK might be an upstream signal for GLT-1 up-regulation induced by sulbactam. We further found that SB203580, the specific inhibitor of p38 MAPK, dose-dependently inhibited the GLT-1 up-regulation induced by sulbactam either in non- or OGD-treated astrocytes and the protective effect of sulbactam on co-cultured neurons against OGD. Taken together, it might be concluded that sulbactam protects cerebral neurons against OGD by up-regulating astrocytic GLT-1 expression via p38 MAPK signal pathway.

11.
Neurochem Res ; 43(9): 1779-1790, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29995175

ABSTRACT

Previous studies have shown that intermittent hypobaric hypoxia (IH) preconditioning protected neurons survival from brain ischemia. However, the mechanism remains to be elucidated. The present study explored the role of nitric oxide (NO) in the process by measuring the expression of NO synthase (NOS) and NO levels. Male Wistar rats (100) were randomly assigned into four groups: sham group, IH + sham group, ischemia group and IH + ischemia group. Rats for IH preconditioning were exposed to hypobaric hypoxia mimicking 5000 m high-altitude (PB = 404 mmHg, PO2 = 84 mmHg) 6 h/day, once daily for 28 days. Global brain ischemia was established by four-vessel occlusion that has been created by Pulsinelli. Rats were sacrificed at 7th day after the ischemia for neuropathological evaluation by thionin stain. In addition, the expression of neuronal NOS (nNOS), inducible NOS (iNOS), and NO content in the hippocampal CA1 subfield were measured at 2nd day and 7th day after the ischemia. Results revealed that global brain ischemia engendered delayed neuronal death (DND), both nNOS and iNOS expression up-regulated, and NO content increased in the hippocampal CA1 subfield. IH preconditioning reduced neuronal injury induced by the ischemia, and prevented the up-regulation of NOS expression and NO production. In addition, L-NAME + ischemia group was designed to detect whether depressing NO production could alleviate the DND. Pre-administration of L-NAME alleviated DND induced by the ischemia. These results suggest that IH preconditioning plays a protective role by inhibiting the over expression of NOS and NO content after brain ischemia.


Subject(s)
Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Hypoxia/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Animals , Brain Ischemia/pathology , CA1 Region, Hippocampal/pathology , Hypoxia/pathology , Male , Random Allocation , Rats , Rats, Wistar
12.
Curr Pharm Des ; 23(33): 5045-5055, 2017.
Article in English | MEDLINE | ID: mdl-28641538

ABSTRACT

Glutamate is the primary excitatory neurotransmitter in the mammalian central nervous system, which plays an important role in many aspects of normal brain function such as neural development, motor functions, learning and memory etc. However, excessive accumulation of glutamate in the extracellular fluid will induce excitotoxicity which is considered to be a major mechanism of cell death in brain ischemia. There is no enzyme to decompose the glutamate in extracellular fluid, so extracellular glutamate homeostasis within the central nervous system is mainly regulated by the uptake activity of excitatory amino acid transporters. Among the five excitatory amino acid transporters, glial glutamate transporter-1 (GLT-1) is responsible for 90% of total glutamate uptake. Thus, GLT-1 is essential for maintaining the appropriate level of extracellular glutamate, and then limiting excitotoxicity of glutamate in central nervous system. Therefore, the regulation of GLT-1 might be a potential therapeutic target for ischemic brain injury. This review summarizes recent advances including our findings in the methods or medicine that could protect neurons against brain ischemic injury via upregulation of GLT-1 and discuss the possible application of these strategies.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Drug Delivery Systems/trends , Glutamate Plasma Membrane Transport Proteins/biosynthesis , Up-Regulation/physiology , Animals , Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Drug Delivery Systems/methods , Estrogen Receptor Modulators/administration & dosage , Excitatory Amino Acid Transporter 2 , Glutamic Acid/metabolism , Histamine Antagonists/administration & dosage , Humans , Up-Regulation/drug effects
13.
Mol Neurobiol ; 54(1): 58-71, 2017 01.
Article in English | MEDLINE | ID: mdl-26732590

ABSTRACT

Our previous study has proved that the up-regulation of glial glutamate transporter 1 (GLT-1) played an important role in the acquisition of brain ischemic tolerance after cerebral ischemic preconditioning (CIP) in rats. However, little is known about the mechanism involved in the up-regulation of GLT-1 in the process. The present study investigates whether p38 MAPK, ERK1/2, and/or JNK participates in the up-regulation of GLT-1 during the induction of brain ischemic tolerance by CIP. It was found that CIP significantly enhanced the expression of p-p38 MAPK without altering p-ERK1/2 and p-JNK expression in the CA1 hippocampus. Inhibition of p38 MAPK function by its selective inhibitor SB203580 or knockdown p38 MAPK expression by its antisense oligodeoxynucleotides (AS-ODNs) suppressed the induction of brain ischemic tolerance. Furthermore, p38 MAPK was activated earlier than the up-regulation of GLT-1 in the CA1 hippocampus after CIP. Meanwhile, the expression of p-p38 MAPK by astrocytes was increased, and p38 MAPK AS-ODNs dose-dependently inhibited the up-regulation of GLT-1 after CIP. Taken together, it could be concluded that p38 MAPK participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance after CIP.


Subject(s)
Brain Ischemia/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Hippocampus/metabolism , Ischemic Preconditioning/methods , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain Ischemia/pathology , Hippocampus/pathology , Male , Rats , Rats, Wistar
14.
Environ Technol ; 38(13-14): 1689-1695, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27776447

ABSTRACT

In this study, the municipal solid waste (MSW) is considered as one kind of energy source in urban planning scheme instead of a trash stream. Considering the characteristics of MSW from different urban functional zones and the current energy supply modes, an evaluation model for waste-to-energy (WtE) method was set up based on the analytical hierarchy process technique. The model consists of three layers: 15 fundamental indices, 4 influencing factors based on fundamental indices and the target functions supported by influencing factors. Taking an urban functional region of a city in north China as the research object, 4 alternatives are compared according to their weights and the sensitivities of the influencing factors are analyzed. The result will provide guide for the disposal method of WtE in new urban district planning and old urban redevelopment.


Subject(s)
Energy-Generating Resources , Models, Theoretical , Refuse Disposal/methods , China , Cities , City Planning
15.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 31(3): 238-43, 2015 May.
Article in Chinese | MEDLINE | ID: mdl-26387186

ABSTRACT

OBJECTIVE: The present study was undertaken to design antisense oligodeoxynucleotides (AS-ODNs) of glial glutamate transporter-la (GLT-1a) and to evaluate the effectiveness of the designed AS-ODNs on the expression of GLT-1a. METHODS: Five sequences of GLT-1a AS-ODNs were designed according to the C terminus specific sequences of GLT-1a mRNA using antisense design software of IDT Com- pany. Western blot analysis was used to evaluate the inhibition effects of the five GLT-1a AS-ODNs on the expression of GLT-la. RESULTS: The sequence of GLT-1a AS-ODNs with sequence of 5'-GGTTCTTCCTCAACACTGCA-3' could specifically inhibit the expression of GLT-1a in the hippocampal CA1 subfield of rats, while it had no effect on the expression of GLT-1b. This sequence showed similar inhibition on the expression of GLT-la in sham and ceftriaxone (Cef)-treated rats. It could also significantly inhibit the cerebral ischemic preconditioning (CIP)-induced up-regulation in the expression of GLT-1a. The magnitude of the inhibition in sham, Cef- or CIP-treated rats was similar by more than 60%. CONCLUSION: From the designed five sequences of GLT-1a AS-ODNs, we obtained an effective sequence which can specifically inhibit the expression of GLT-1a.


Subject(s)
CA1 Region, Hippocampal/metabolism , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Oligonucleotides, Antisense/genetics , Animals , Excitatory Amino Acid Transporter 2/metabolism , Ischemic Preconditioning , RNA, Messenger , Rats , Up-Regulation
16.
J Neurochem ; 132(2): 194-205, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25270764

ABSTRACT

Ceftriaxone(Cef) selectively increases the expression of glial glutamate transporter-1 (GLT-1), which was thought to be neuroprotective in some circumstances. However, the effect of Cef on glutamate uptake of GLT-1 was mostly assayed using in vitro studies such as primary neuron/astrocyte cultures or brain slices. In addition, the effect of Cef on neurons in different ischemic models was still discrepant. Therefore, this study was undertaken to observe the effect of Cef on neurons in global brain ischemia in rats, and especially to provide direct evidence of the up-regulation of GLT-1 uptake for glutamate contributing to the neuronal protection of Cef against brain ischemia. Neuropathological evaluation indicated that administration of Cef, especially pre-treatment protocols, significantly prevented delayed neuronal death in hippocampal CA1 subregion normally induced by global brain ischemia. Simultaneously, pre-administration of Cef significantly up-regulated the expression of GLT-1. Particularly, GLT-1 uptake assay with (3) H-glutamate in living cells from adult rats showed that up-regulation in glutamate uptake accompanied up-regulated GLT-1 expression. Inhibition of GLT-1 by antisense oligodeoxynucleotides or dihydrokainate significantly inhibited the Cef-induced up-regulation in GLT-1 uptake and the neuroprotective effect against global ischemia. Thus, we may conclude that Cef protects neurons against global brain ischemia via up-regulation of the expression and glutamate uptake of GLT-1. Glutamate uptake by glial glutamate transporter-1 (GLT-1) is the principal way to regulate extracellular glutamate homeostasis in central nervous system. Over-accumulation of glutamate results in excitotoxicity and injures neurons after cerebral ischemia. Ceftriaxone up-regulates GLT-1 expression and uptake of glutamate, diminishes the excitotoxicity of glutamate and then protects neurons against global brain ischemia.


Subject(s)
Brain Ischemia/drug therapy , Ceftriaxone/therapeutic use , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Neuroprotective Agents/therapeutic use , Animals , Biological Transport/drug effects , Brain Ischemia/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Ceftriaxone/administration & dosage , Ceftriaxone/pharmacology , Drug Evaluation, Preclinical , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Knockdown Techniques , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Random Allocation , Rats , Rats, Wistar , Up-Regulation
17.
Mol Neurobiol ; 51(3): 1322-33, 2015.
Article in English | MEDLINE | ID: mdl-25064054

ABSTRACT

The study was undertaken to investigate whether sulbactam protects cerebral neurons against ischemia and whether the protection is mediated by regulating the expression and uptake activity of glial glutamate transporter-1 (GLT1) in a rat global brain ischemia model. The CA1 hippocampus was selected as the observing target. Real time quantitative PCR, Western blot and immunohistochemistry assays were used to detect GLT1 expression. Neuropathological evaluation was performed after thionin staining to determine the extent of the delayed neuronal death (DND) of pyramidal neurons. It was found that cerebral ischemia for 8 min induced obvious DND of pyramidal neurons and GLT1 downregulation. Sulbactam pretreatment significantly upregulated GLT1 expression in sham rats and prevented or reversed the GLT1 downregulation normally induced in the ischemic rat brain. Meanwhile, sulbactam pretreatment effectively prevented the DND of pyramidal neurons normally induced by brain ischemia in a dose-dependent manner. Sulbactam posttreatment also protected pyramidal neurons against DND induced by brain ischemia although the magnitude of the protective effect was weaker than that in sulbactam pretreatment. Furthermore, either antisense knockdown of GLT1 expression or inhibition of the GLT1 uptake activity with dihydrokainate, a selective inhibitor of GLT1, significantly blocked the neuronal protective effect of sulbactam. These findings indicate that sulbactam has a neuronal protective effect though upregulating GLT1.


Subject(s)
Brain Ischemia/drug therapy , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/drug effects , Neurons/drug effects , Sulbactam/pharmacology , Animals , Brain Ischemia/metabolism , Cell Death/drug effects , Cell Death/physiology , Hippocampus/metabolism , Ischemic Preconditioning/methods , Male , Neurons/metabolism , Rats, Wistar , Up-Regulation
18.
Mol Neurobiol ; 47(1): 197-208, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23180278

ABSTRACT

Recent evidence suggests that limb ischemic preconditioning (LIP) protects neurons against cerebral ischemia-reperfusion injury. However, the mechanisms of LIP are not well understood. Neuroglobin (Ngb) is a recently discovered globin that affords protection against hypoxic/ischemic brain injury. This study was performed to investigate the role of Ngb in the neuroprotection of LIP against brain ischemia and the involvements of mitochondria in the process. The rat global brain ischemic model was used, and the CA1 hippocampus was selected as the observational target. Ngb expression was investigated by RT-PCR and Western blot. Neuropathological evaluation was performed by thionin staining. Mitochondrial membrane potential (Δψm), Na(+)-K(+)-ATPase activity, and ultrastructure were examined by flow cytometry, spectrophotometry, and transmission electron microscopy, respectively. We also used Ngb antisense oligodeoxynucleotides (AS-ODNs) and Ngb inducer hemin to inhibit or mimic the effect of LIP. We found that LIP significantly up-regulated Ngb expression and protected neurons against ischemia. Furthermore, LIP effectively improved deterioration in the Δψm, mitochondrial Na(+)-K(+)-ATPase activity, and ultrastructure induced by cerebral ischemia. These effects of LIP were inhibited partly by Ngb AS-ODNs and mimicked by hemin. It could be concluded that up-regulation of Ngb expression played an important role in the neuroprotection induced by LIP, and the Ngb-mediated neuroprotection of LIP was, at least partly, associated with mitochondria.


Subject(s)
Extremities/blood supply , Extremities/pathology , Globins/metabolism , Ischemic Preconditioning , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/metabolism , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Globins/genetics , Hemin/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Nerve Tissue Proteins/genetics , Neuroglobin , Oligonucleotides, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Up-Regulation/drug effects , Up-Regulation/genetics
19.
Article in Chinese | MEDLINE | ID: mdl-22737922

ABSTRACT

OBJECTIVE: To investigate whether formalin inflammatory pain can induce the change in heme oxygenase-1 (HO-1) expression in the spinal cord of rats or not and the time course character. METHODS: 42 SD rats were divided into 7 groups (n = 6): control formalin 6 h, formalin 12 h, formalin 1 d, formalin 2 d, formalin 3 d and formalin 7 d groups. Rats were subcutaneously injected with 0.2 ml 0.5% formalin into the ventral surface of right hind paw to induce periphery inflammatory pain. The immunohistochemistry was used to observe the expression of HO-1 protein in laminae I - II of the spinal cord dorsal horn and the area around canalis centralis of the I5 spinal segment of rats. RESULTS: There are rare HO-1 immunoreactive cells in the laminae I - II of the dorsal horn and the area around canalis centralis of the I5 spinal segment of rats of control group and HO-1 immunoreactive cells were light in staining degree. Comparing with control group, the numbers of HO-1 immunoreactive cells in the I - II laminae of dorsal horn and area around canalis centralis were increased in the rats at 6 h after formalin injection. The number and staining degree of HO-1 immunoreactive cells were further increased at 12 h and peaked at 1 d after formalin injection. They didn't return to normal level at the 7th day. There were no difference in right and left dorsal horn in the number and staining degree of HO-1 immunoreactive cells at the same time after formalin injection. CONCLUSION: Formalin inflammatory pain induced increased expression of HO-1 in the spinal cord dorsal horn and the area around canalis centralis of rats. At 1 d after injection of formalin, the increased expression of HO-1 was the most obviously.


Subject(s)
Formaldehyde/adverse effects , Heme Oxygenase (Decyclizing)/metabolism , Spinal Cord/metabolism , Animals , Male , Pain/metabolism , Pain Measurement , Rats , Rats, Sprague-Dawley
20.
Acta Pharmacol Sin ; 33(7): 897-908, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22659625

ABSTRACT

AIM: Plasminogen activator inhibitor-1 (PAI-1) is involved in the progression of pulmonary fibrosis. The present study was undertaken to examine the effects on pulmonary fibrosis of silencing PAI-1 expression with small interfering RNA (siRNA) and to assess the possible underlying mechanisms. METHODS: Male Wistar rats were subjected to intratracheal injection of bleomycin (BLM, 5 mg/kg, 0.2 mL) to induce pulmonary fibrosis. Histopathological changes of lung tissue were examined with HE or Masson's trichrome staining. The expression levels of α-smooth muscle actin (α-SMA), collagen type-I and type-III, caspase-3, as well as p-ERK1/2 and PI3K/Akt in the lung tissue were evaluated using imunohistochemistry and Western blot analyses. The fibroblasts isolated from BLM-induced fibrotic lung tissue were cultured and transfected with pcDNA-PAI-1 or PAI-1siRNA. The expression level of PAI-1 in the fibroblasts was measured using real time RT-PCR and Western blot analysis. The fibroblast proliferation was evaluated using MTT assay. RESULTS: Intratracheal injection of PAI-1-siRNA (7.5 nmoL/0.2 mL) significantly alleviated alveolitis and collagen deposition, reduced the expression of PAI-1, α-SMA, collagen type-I and collagen type-III, and increased the expression of caspase-3 in BLM-induced fibrotic lung tissue. In consistence with the in vivo results, the proliferation of the cultured fibroblasts from BLM-induced fibrotic lung tissue was inhibited by transfection with PAI-1-siRNA, and accelerated by overexpression of PAI-1 by transfection with pcDNA-PAI-1. The expression of caspase-3 was increased as a result of PAI-1 siRNA transfection, and decreased after transfection with pcDNA-PAI-1. In addition, the levels of p-ERK1/2 and PI3K/Akt in the fibrogenic lung tissue were reduced after treatment with PAI-1siRNA. CONCLUSION: The data demonstrate that PAI-1 siRNA inhibits alveolitis and pulmonary fibrosis in BLM-treated rats via inhibiting the proliferation and promoting the apoptosis of fibroblasts. Suppression ERK and AKT signalling pathways might have at least partly contributed to this process. Targeting PAI-1 is a promising therapeutic strategy for pulmonary fibrosis.


Subject(s)
Plasminogen Activator Inhibitor 1/genetics , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Actins/analysis , Actins/metabolism , Animals , Apoptosis , Bleomycin , Caspase 3/metabolism , Cells, Cultured , Collagen/analysis , Collagen/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Hydroxyproline/analysis , Hydroxyproline/metabolism , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System , Male , Phosphatidylinositol 3-Kinase/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...