Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Chem Biol Interact ; 368: 110206, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36195188

ABSTRACT

Growing pieces of evidence suggest that Alzheimer's disease (AD) is interlinked with Type 2 diabetes mellitus (DM), which has been described as "type 3 DM". In this study, we investigate the neuronal insult attributable to advanced glycation end products (AGEs) as the models of DM-related AD to understand the effects exerted by calycosin on neurodegenerative changes both in vivo and in vitro studies and also studied the associated molecular mechanisms. The results reported herein revealed that the viability of the PC12 cells induced by AGEs increased when treated with calycosin. It was also observed that the learning and memory abilities of AGE-induced DM-related AD rats improved under these conditions. Analysis of the reported results indicates that calycosin can effectively down-regulate the activity of GSK-3ß to result in the reversal of the process of tau hyperphosphorylation, inhibit the expression of RAGE and BACE-1 proteins, resulting in a decrease in the production of ß-amyloid and regulate the PGC-1α/TFAM signaling pathway to repair mitochondrial dysfunction. It can be inferred that calycosin can potentially exhibit important therapeutic properties that can be exploited during the treatment of AD, especially DM-related AD.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Animals , Rats , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Glycation End Products, Advanced/metabolism , Glycogen Synthase Kinase 3 beta , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism
3.
Molecules ; 27(15)2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35956985

ABSTRACT

Alzheimer's disease (AD) is a major neurodegenerative disease, but so far, it can only be treated symptomatically rather than changing the process of the disease. Recently, triazoles and their derivatives have been shown to have potential for the treatment of AD. In this study, the neuroprotective effects of 4-(4-(heptyloxy)phenyl)-2,4-dihydro-3H-1,2,4-triazol-3-one (W112) against ß-amyloid (Aß)-induced AD pathology and its possible mechanism were explored both in vitro and in vivo. The results showed that W112 exhibits a neuroprotective role against Aß-induced cytotoxicity in PC12 cells and improves the learning and memory abilities of Aß-induced AD-like rats. In addition, the assays of the protein expression revealed that W112 reversed tau hyperphosphorylation and reduced the production of proinflammatory cytokines, tumor necrosis factor-α and interleukin-6, both in vitro and in vivo studies. Further study indicated that the regulation of mitogen-activated protein kinase/nuclear factor-κB pathways played a key role in mediating the neuroprotective effects of W112 against AD-like pathology. W112 may become a potential drug for AD intervention.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Neuroprotective Agents , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , MAP Kinase Signaling System , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Rats
5.
Molecules ; 27(12)2022 Jun 18.
Article in English | MEDLINE | ID: mdl-35745036

ABSTRACT

Advanced glycation end products (AGEs) are stable products produced by the reaction of macromolecules such as proteins, lipids or nucleic acids with glucose or other reducing monosaccharides, which can be identified by immunohistochemistry in the senile plaques and neurofibrillary tangles of Alzheimer's disease (AD) patients. Growing evidence suggests that AGEs are important risk factors for the development and progression of AD. 1,8-cineole (CIN) is a monoterpenoid compound which exists in many plant essential oils and has been proven to have neuroprotective activity, but its specific effect and molecular mechanisms are not clear. In this study, AGEs-induced neuronal injury and intracerebroventricular-AGE animals as the possible models for AD were employed to investigate the effects of CIN on AD pathology as well as the molecular mechanisms involved both in vivo and in vitro. Our study demonstrated that CIN could ameliorate tau phosphorylation by down-regulating the activity of GSK-3ß and reducing Aß production by inhibiting the activity of BACE-1 both in vivo and in vitro. It is suggested that CIN has certain therapeutic value in the treatment of AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Eucalyptol/pharmacology , Glycation End Products, Advanced/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Phosphorylation , tau Proteins/metabolism
6.
Am J Transl Res ; 13(8): 8885-8896, 2021.
Article in English | MEDLINE | ID: mdl-34540002

ABSTRACT

OBJECTIVE: Alzheimer's disease (AD) is a common neurodegenerative disease. This study was designed to investigate the roles of lncRNA NEAT1/miR-27a-3p axis in AD. METHODS: Amyloid protein was used to treat SH-SY5Y cells and rats to construct AD model. RT-qPCR was used to quantify lncRNA NEAT1 and micro-27a-3p in AD model cells. Western blot was used to determine the ß-amyloid-precursor-protein-cleaver-enzyme 1 (BACE1), amyloid, Tau protein and its phosphorylation, Caspase 3 protein and its lytic cell protein and amyloid precursor protein (APP). Flow cytometry was used to detect apoptosis. The cell activity was detected by CCK-8. The lncRNA NEAT1 and miR-27a-3p inhibition or over-expression vectors were constructed. The dual luciferase reporter gene and RNA pull-down assay were used to detect the targeting relationship between lncRNA NEAT1 and micro-27a-3p. The cognitive function of rats was tested by water maze. RESULTS: After being induced by amyloid protein, lncRNA NEAT1 was up-regulated while micro-27a-3p was down-regulated in SH-SY5Y cells. Apoptosis rate was increased and cell activity was decreased. Amyloid protein, BACE1 protein, APP protein, Tau protein and its phosphorylation, Caspase 3 protein and its lytic cell protein were up-regulated. Down-regulation of lncRNA NEAT1 or up-regulation of micro-27a-3p could reduce cell apoptosis, increase cell activity, down-regulate amyloid protein, BACE1 protein, APP protein, Tau protein and its phosphorylation, and up-regulate caspase 3 protein and its lysate protein. Dual luciferase reporter gene assay and RNA pull-down experiments revealed that micro-27a-3p was the target gene of lncRNA NEAT1. Down-regulation of micro-27a-3p could offset the changes caused by LncRNA NEAT1. AD caused cognitive dysfunction in rats, which was improved by down-regulation of lncRNA NEAT1. CONCLUSION: lncRNA NEAT1 regulates the development of AD by down-regulating micro-27a-3p.

7.
Brain Res Bull ; 175: 254-262, 2021 10.
Article in English | MEDLINE | ID: mdl-34217799

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief as there are concerns about the reliability of the results. Concerns have been raised about the western blot bands in Figures 6 B + D having the same eyebrow shaped phenotype as found in many other publications as detailed here (https://pubpeer.com/publications/B32F93859FBAA13471ED0FFCA5BCB6). The journal requested the corresponding author to comment on these concerns and send the raw data, however the author was not able to provide uncropped images of the original gels. The Editor-in-Chief therefore no longer has confidence in the data and conclusions of this study.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apoptosis/genetics , Autophagy/genetics , Hippocampus/pathology , Immediate-Early Proteins/genetics , MicroRNAs/genetics , Neurons/pathology , Tumor Suppressor Proteins/genetics , Alzheimer Disease/psychology , Animals , Cell Survival/genetics , Cells, Cultured , Maze Learning , Mice , Mice, Transgenic
8.
Int Immunopharmacol ; 97: 107578, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33892301

ABSTRACT

OBJECTIVE: Evidences have indicated that interleukin-1ß (IL-1ß) and microRNAs (miRNAs) are implicated in Alzheimer's disease (AD), and we aimed to study the role of IL-1ß in AD development with the involvement of miR-9-5p and targeting protein for xenopus kinesin-like protein 2 (TPX2). METHODS: APPswe/PS1dE9 double transgenic mice and C57BL/6 wild type mice were treated with inhibited IL-1ß, miR-9-5p mimic and/or silenced TPX2. Expression of IL-1ß, miR-9-5p, TPX2, amyloid-ß (Aß) and p-tau in mouse hippocampal tissues was determined. The behavioral changes, hippocampal pathological injury, Aß plaque deposition, tau expression, neuronal apoptosis and oxidative stress of AD mice were all measured. The regulatory relationships between IL and 1ß and miR-9-5p, and between miR-9-5p and TPX2 were confirmed. RESULTS: IL-1ß and TPX2 were upregulated while miR-9-5p was downregulated in hippocampal tissues from AD mice versus non-transgenic littermate mice. Inhibited IL-1ß or elevated miR-9-5p improved behavioral changes and neuronal injury of AD mice, and suppressed plaque deposition and oxidative stress in hippocampal tissues of AD mice. These changes that induced by elevated miR-9-5p could be reversed by overexpression of TPX2. IL-1ß negatively regulated miR-9-5p, and TPX2 was a target gene of miR-9-5p. CONCLUSION: This study suggested that inhibition of IL-1ß played a protective role in AD by promoting miR-9-5p and downregulating TPX2, which may contribute to exploration on AD treatment.


Subject(s)
Alzheimer Disease/immunology , Cell Cycle Proteins/genetics , Interleukin-1beta/metabolism , MicroRNAs/metabolism , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Cycle Proteins/metabolism , Disease Models, Animal , Down-Regulation/immunology , Female , Humans , Mice , Mice, Transgenic , MicroRNAs/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Presenilin-1/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/immunology
9.
J Ethnopharmacol ; 273: 114002, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33705924

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The traditional Chinese medicine Sanweidoukou decoction (DK-3) was a classical formula for the treatment of nervous system diseases, recorded in the Chinese medical classic Sibu Yidian. AIM OF THE STUDY: The present study is aim to investigate the neuroprotective effects of DK-3 on ß-amyloid (Aß) protein -induced AD-like pathologies and underlying molecular mechanisms both in vitro and in vivo studies. MATERIALS AND METHODS: Hydrolysates of DK-3 were analyzed by LC-ESI-MS/MS. In vitro, MTT was utilized to examine effects of DK-3 on Aß25-35-induced cytotoxicity in PC12 cells. In vivo, male Sprague-Dawley rats were administered with Aß25-35 to induce AD-like pathologies and behavioral evaluations were conducted via Morris water maze (MWM) test. Histopathological changes were observed by Hematoxylin-eosin (HE) straining. Immunohistochemistry (IHC) was used to detect the tau hyperphosphorylation at Thr181 site. The expression levels of tau hyperphosphorylation, inflammation-related cytokines such as COX-2, iNOS, TNF-α, IL-1ß, IL-6, the phosphorylated state of various mitogen-activated protein kinase (MAPK) signaling molecules (p38 MAPK, ERK, and JNK) and activation of nuclear factor κB (NF-κB) in vitro and in vivo were assessed via Western blot. RESULTS: In vitro, DK-3 dose-dependently increased cell viability of PC12 cells induced by Aß25-35. In vivo, DK-3 improved learning and memory abilities of Aß25-35-induced AD-like rats. Moreover, DK-3 reversed hyperphosphorylation of tau and reduced the production of inflammation-related cytokines through significantly inhibited MAPK and NF-κB signaling pathways both in vitro and in vivo studies. CONCLUSION: The present study suggested that the traditional Chinese medicine DK-3 may play a role in preventing and treating AD by reducing the hyperphosphorylation of tau protein and the expressions of inflammation-related cytokines via modulating the MAPK/NF-κB signaling pathways.


Subject(s)
Amyloid beta-Peptides/toxicity , Drugs, Chinese Herbal/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Neurons/drug effects , Peptide Fragments/toxicity , Animals , Cell Survival/drug effects , Chromatography, Liquid , Gene Expression Regulation/drug effects , Male , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/genetics , PC12 Cells , Phytotherapy , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction , Tandem Mass Spectrometry/methods
10.
Cell Cycle ; 20(1): 112-125, 2021 01.
Article in English | MEDLINE | ID: mdl-33410377

ABSTRACT

Accumulating data manifest that long non-coding RNA (lncRNAs) are involved in all kinds of neurodegenerative disorders, consisting of the onset and progression of Alzheimer's disease (AD). The study was for the research of the mechanism of lncRNA H19 (H19) in viability and apoptosis of PC12 cells induced by Aß25-35 in a cellular model of AD with the regulation of microRNA (miR)-129 and high mobility group box-1 protein (HMGB1). An AD cellular model of PC12 cells was established using Aß25-35. The Aß25-35-induced PC12 cells were transfected with si-H19 or miR-129 mimic to figure their roles in cell viability,apoptosis, mitochondrial membrane potential dysfunction and oxidative stress in AD. Luciferase reporter assay and RNA-pull down assay were employed for verification of the binding relationship between H19 and miR-129 and the targeting relationship between miR-129 and HMGB1. An AD mouse model was induced and brain tissues were collected. H19, miR-129 and HMGB1 were detected in Aß25-35-treated cells and brain tissues of AD mice. Elevated H19, HMGB1 and decreased miR-129 were found in Aß25-35-treated PC12 cells as well as in brain tissues of AD mice. Silenced H19 or elevated miR-129 promoted viability, inhibited apoptosis, prevented mitochondrial membrane potential dysfunction and decreased oxidative stress in Aß25-35-treated PC12 cells. H19 could specifically bind to miR-129. MiR-129 specifically suppressed HMGB1 expression. This study suggests that silenced H19 and up-regulated miR-129 accelerates viability and represses apoptosis of PC12 cells stimulated by Aß25-35 in AD, which is beneficial for AD treatment.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Apoptosis/genetics , Cell Survival/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Up-Regulation/genetics , Animals , Cell Line, Tumor , HMGB1 Protein/genetics , Membrane Potential, Mitochondrial/genetics , Neurons/pathology , Oxidative Stress/genetics , PC12 Cells , Peptide Fragments/genetics , Rats , Signal Transduction/genetics
11.
Cell Physiol Biochem ; 51(3): 1399-1409, 2018.
Article in English | MEDLINE | ID: mdl-30485839

ABSTRACT

BACKGROUND/AIMS: Temporal lobe epilepsy (TLE) is the most common form of adult localization-related epilepsy that is accompanied by progressive etiopathology and high incidences of drug resistance. Circular RNAs (circRNAs) play important roles in fine-tuning gene expression, however, the expression profile and clinical significance of circRNAs in TLE remains unknown. METHODS: Circular RNA microarray was conducted to identify TLE-related circRNAs. CCK8 assays and flow cytometric assays were conducted to clarify the role of circRNA in TLE in vitro. Bioinformatics analysis and in vitro experiments were conducted to clarify the mechanism of circRNA-mediated gene regulation in TLE cell. RESULTS: 586 differentially expressed circRNAs were identified between TLE and the control tissues. The expression of circRNA-0067835 was significantly down-regulated in tissues and plasma from TLE patients. Lower circRNA-0067835 correlated to increased seizure frequency, HS, and higher Engel's score. Overexpression of circRNA-0067835 observably decreased SH-SY5Y cell proliferation by causing G1 arrest and promoting apoptosis. Bioinformatics online programs predicted that circRNA-0067835 acted as miR-155 sponge to regulate FOXO3a expression, which was validated using luciferase reporter assay. CONCLUSION: Our experiments showed that circRNA-0067835 regulated refractory epilepsy progression by acting as a sponge of miR-155 to promote FOXO3a expression, indicating that circRNA-0067835 may serve as a potential therapeutic target for patients with TLE.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Gene Expression Profiling , Gene Expression Regulation , MicroRNAs/genetics , RNA/genetics , Cell Line , Cell Proliferation , Epilepsy, Temporal Lobe/pathology , Forkhead Box Protein O3/genetics , Humans , RNA, Circular
12.
Oncotarget ; 9(37): 24871, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29873327

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.23119.].

13.
Oncotarget ; 9(9): 8542-8547, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29492215

ABSTRACT

Mesial temporal lobe epilepsy (mTLE), the most common type of temporal lobe epilepsy (TLE), is particularly relevant due to its high frequency of therapeutic resistance of anti-epileptic therapies. MicroRNAs (miRNAs) have been shown to be dysregulated in epilepsy and neurodegenerative diseases, and we hypothesized that miRNAs could be involved in the pathogenesis of MTLE. The present study aimed to explore the expression and functions of miRNA-153 in mTLE. The expression levels of miRNA-153 in refractory TLE patients were evaluated. The bioinformatics analysis showed that the potential target genes of miR-153 were involved in biological processes, molecular functions, and cellular components. miRNA-153 is significantly dysregulated in temporal cortex and plasma of mTLE patients. We identify HIF-1α as a direct target of miRNA-153, and luciferase reporter assays demonstrated that miR-153 could regulate the HIF-1αexpression via 3'-UTR pairing. These data suggest that miR-153 might represent a useful biomarker and treatment target for patients with mTLE.

14.
Oncotarget ; 8(53): 91551-91557, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29207665

ABSTRACT

Beta-site Amyloid precursor protein Cleaving Enzyme 1 (BACE1) is conceived as a potential target for therapies against Alzheimer disease (AD). MicroRNAs (miRNAs) are small non-coding RNAs that negatively regulate gene expression in a sequence-specific manner. Although miRNAs have been increasingly recognized as important modulators in sporadic AD. In order to confirm whether miR-15b correlates with the BACE1 upregulation in sporadic AD, we firstly evaluated the expression of miR-15b and BACE1 in sporadic AD brain tissues and analyzed the correlation of miR-15b with BACE1. Then we determined the regulation of miR-15b in SH-SY5Y cells on the BACE1 expression. And finally we determined the targeting to 3' UTR of BACE1 by miR-15b by a luciferase reporter. Downregulation of miR-15b alleviated Aß-induced viability inhibition and decreased apoptosis in SH-SY5Y cells. Our results demonstrated that miR-15b play an important role in the cellular AD phenotype and might be involved in the pathogenesis of AD.

15.
Oncotarget ; 8(69): 114065-114071, 2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29371969

ABSTRACT

Although large numbers of microRNAs (miRNAs) expressed in Alzheimer disease (AD) have been detected, their functions and mechanisms of regulation remain to be fully clarified. Beta-site Amyloid precursor protein Cleaving Enzyme 1 (BACE1) has been one of the prime therapeutic targets for AD. Here, we identified that miR-124 levels are gradually decreased in AD. In addition, we demonstrated that miR-124 suppresses BACE1 expression by directly targeting the 3'UTR of Bace1 mRNA in vitro. Inhibition of miR-124 significantly increased BACE1 levels in neuronal cells. In contrast, miR-124 overexpression significantly suppressed BACE1 expression in cells. And finally we determined that downregulation of miR-124 alleviated Aß-induced viability inhibition and decreased apoptosis in SH-SY5Y cells. Our results demonstrated that miR-124 is a potent negative regulator of BACE1 in the cellular AD phenotype and might be involved in the pathogenesis of AD.

16.
Neurosci Lett ; 612: 193-198, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26679229

ABSTRACT

Our previous study has demonstrated that glucagon-like peptide-1 (GLP-1) receptor agonist could protect neurons from advanced glycation end products (AGEs) toxicity in vitro. However, further studies are still needed to clarify the molecular mechanism of this GLP-1 receptor -dependent action. The present study mainly focused on the effect of GLP-1 receptor agonists against the receptor for advanced glycation end products (RAGE) signal pathway and the mechanism underlying this effect of GLP-1. Firstly the data based on the SH-GLP-1R(+) and SH-SY5Y cells confirmed our previous finding that GLP-1 receptor could mediate the protective effect against AGEs. The assays of the protein activity and of the mRNA level revealed that apoptosis-related proteins such as caspase-3, caspase-9, Bax and Bcl-2 were involved. Additionally, we found that both GLP-1 and exendin-4 could reduce AGEs-induced reactive oxygen species (ROS) accumulation by suppressing the activity of nicotinamide adenine dinucleotide phosphate-oxidase. Interestingly, we also found that GLP-1 receptor activation could attenuate the abnormal expression of the RAGE in vitro and in vivo. Furthermore, based on the analysis of the protein expression and translocation level of transcription factor nuclear factor-κB (NF-κB), and the use of GLP-1 receptor antagonist exendin(9-39) and NF-κB inhibitor pyrrolidine dithiocarbamate, we found that the effect mediated by GLP-1 receptor could alleviate the over expression of RAGE induced by ligand via the suppression of NF-κB. In summary, the results indicated that inhibiting RAGE/oxidative stress was involved in the protective effect of GLP-1 on neuron cells against AGEs induced apoptosis.


Subject(s)
Apoptosis , Glucagon-Like Peptide-1 Receptor/metabolism , Glycation End Products, Advanced/metabolism , Neurons/cytology , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Animals , Cell Line, Tumor , Exenatide , Glucagon-Like Peptide-1 Receptor/agonists , Glycation End Products, Advanced/toxicity , Humans , Male , Mice, Inbred ICR , NADPH Oxidases/metabolism , NF-kappa B/pharmacology , Neurons/metabolism , Peptides/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Venoms/metabolism
17.
Age (Dordr) ; 34(5): 1211-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21901364

ABSTRACT

Growing evidence suggests that type 2 diabetes mellitus (DM) is associated with age-dependent Alzheimer's disease (AD), the latter of which has even been considered as type 3 diabetes. Several physiopathological features including hyperglycemia, oxidative stress, and dysfunctional insulin signaling relate DM to AD. In this study, high glucose-, oxidative stress-induced neuronal injury and intracerebroventricular-streptozotocin (ICV-STZ) animals as the possible models for diabetes-related AD were employed to investigate the effects of exendin-4 (Ex-4), a long-acting glucagon-like peptide-1 (GLP-1) receptor agonist, on diabetes-associated Alzheimer-like changes as well as the molecular mechanisms involved. Our study demonstrated that GLP-1/Ex-4 could exert a protective effect against reduced viability of PC12 cells caused by high glucose and that this protective effect was mediated via the PI3-kinase pathway. In addition, GLP-1/Ex-4 ameliorated oxidative stress-induced injury in PC12 cells. In rat models, bilateral ICV-STZ administration was used to produce impaired insulin signaling in the brain. Fourteen days following ICV-STZ injection, rats treated with twice-daily Ex-4 had better learning and memory performance in the Morris water maze test compared with rats treated with saline. Additionally, histopathological evaluation confirmed the protective effects of Ex-4 treatment on hippocampal neurons against degeneration. Furthermore, we demonstrated that Ex-4 reversed ICV-STZ-induced tau hyperphosphorylation through downregulation of GSK-3ß activity, a key kinase in both DM and AD. Our findings suggests that Ex-4 can protect neurons from diabetes-associated glucose metabolic dysregulation insults in vitro and from ICV-STZ insult in vivo, and that Ex-4 may prove of therapeutic value in the treatment of AD especially DM-related AD.


Subject(s)
Alzheimer Disease/drug therapy , Diabetes Mellitus, Experimental/complications , Hippocampus/pathology , Nerve Degeneration/prevention & control , Oxidative Stress , Peptides/therapeutic use , Venoms/therapeutic use , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Blood Glucose/metabolism , Cell Survival , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Exenatide , Glucagon-Like Peptide 1 , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Male , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...