Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 161: 114474, 2023 May.
Article in English | MEDLINE | ID: mdl-36878051

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease mainly characterized by progressive cognitive dysfunction and memory impairment. Recent studies have shown that regulating silent information regulator 1 (SIRT1) expression has a significant neuroprotective effect, and SIRT1 may become a new therapeutic target for AD. Natural molecules are an important source of drug development for use in AD therapy and may regulate a wide range of biological events by regulating SIRT1 as well as other SIRT1-mediated signaling pathways. This review aims to summarize the correlation between SIRT1 and AD and to identify in vivo and in vitro studies investigating the anti-AD properties of natural molecules as modulators of SIRT1 and SIRT1-mediated signaling pathways. A literature search was conducted for studies published between January 2000 and October 2022 using various literature databases, including Web of Science, PubMed, Google Scholar, Science Direct, and EMBASE. Natural molecules, such as resveratrol, quercetin, icariin, bisdemethoxycurcumin, dihydromyricetin, salidroside, patchouli, sesamin, rhein, ligustilide, tetramethoxyflavanone, 1-theanine, schisandrin, curcumin, betaine, pterostilbene, ampelopsin, schisanhenol, and eriodictyol, have the potential to modulate SIRT1 and SIRT1 signaling pathways, thereby combating AD. The natural molecules modulating SIRT1 discussed in this review provide a potentially novel multi-mechanistic therapeutic strategy for AD. However, future clinical trials need to be conducted to further investigate their beneficial properties and to determine the safety and efficacy of SIRT1 natural activators against AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Sirtuin 1/metabolism , Resveratrol/therapeutic use , Memory Disorders
2.
PeerJ ; 10: e14538, 2022.
Article in English | MEDLINE | ID: mdl-36530401

ABSTRACT

Background: This study examined the association between type 2 diabetes mellitus (T2DM) and 5-year overall survival (OS) in patients with pancreatic cancer (PC). Methods: This retrospective cohort study included patients diagnosed with stage I/II PC at Shengjing Hospital of China Medical University from January 2012 to December 2017. All patients had pancreatic ductal adenocarcinoma or its subtypes. The outcome was the 5-year OS rate based on data from the patient charts. Data analysis was performed using SPSS 22.0. Results: A total of 238 patients were included: 72 with T2DM and 166 without T2DM. There were significant differences in blood glucose levels and OS between the two groups (all P < 0.05). The median OS was 11.4 (95% confidence interval CI [8.49-14.31]) months in the T2DM group and 16.3 (95% CI [12.44-20.16], P = 0.023) months in the non-T2DM group. After adjustment for confounders, T2DM was an independent factor affecting 5-year OS (P = 0.010). Compared with non-T2DM patients, T2DM patients had a higher risk of death (HR = 1.475, 95% CI [1.096-1.985]). Conclusions: T2DM is associated with 5-year OS in patients with PC.


Subject(s)
Carcinoma, Pancreatic Ductal , Diabetes Mellitus, Type 2 , Pancreatic Neoplasms , Humans , Diabetes Mellitus, Type 2/complications , Retrospective Studies , Pancreatic Neoplasms/epidemiology , Carcinoma, Pancreatic Ductal/complications , Pancreatic Hormones , Pancreatic Neoplasms
3.
Brain Res Bull ; 183: 142-152, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35289299

ABSTRACT

Sevoflurane, a commonly used inhaled anesthetic, causes endogenous apoptosis in fetal rats. Microglia polarization is associated with inflammation, and the IL-10/STAT3/SOCS3 pathway is involved in this process. Neuroglobin (Ngb) is a neuroprotective protein which exhibits an anti-inflammatory effect. The purpose of this study was to investigate whether neurotoxicity induced by sevoflurane exposure in prenatal rats correlates with neuroinflammation and microglia polarization and whether Ngb can moderate this response. We found that exposure to sevoflurane on the 20th day of gestation (G20) induced discernable inflammation in postnatal day 0 (P0) rats, promoted M1 polarization of microglia, and inhibited M2 polarization. Hemin-mediated Ngb elevation inhibited sevoflurane-induced neuroinflammation. Additionally, elevated Ngb inhibited M1 polarization and promoted M2 polarization of microglia. We also found that elevated Ngb could alleviate the effect of sevoflurane on the expression of Interleukin-10 (IL-10), phosphorylated-signal transduction and activators of transcription 3 (P-STAT3), and suppressor of cytokine signaling 3 (SOCS3). Furthermore, we found that elevated Ngb ameliorated the effects of sevoflurane on long-term exploratory behavior and learning and memory in the offspring. Our results show that Ngb alleviates the neurotoxicity of sevoflurane to fetal rats by inhibiting neuroinflammation and affecting microglial polarization, a process which may involve the IL-10/STAT3/SOCS3 pathway.


Subject(s)
Microglia , Neurotoxicity Syndromes , Animals , Female , Microglia/metabolism , Neuroglobin/metabolism , Neuroglobin/pharmacology , Neuroinflammatory Diseases , Neurotoxicity Syndromes/metabolism , Pregnancy , Rats , Sevoflurane/pharmacology
4.
Mediators Inflamm ; 2020: 5458061, 2020.
Article in English | MEDLINE | ID: mdl-32655312

ABSTRACT

To investigate the mechanism dexmedetomidine in relieving the neurotoxicity of a developing brain induced by sevoflurane. Sprague-Dawley rats, 6 days old, were randomly divided into three groups. Rats in the control group were inhaled with air after injection of normal saline; rats in the sevoflurane group were injected with normal saline and inhaled with 3% sevoflurane for 2 h in three consecutive day; rats in the dexmedetomidine group were inhaled with 3% sevoflurane after intraperitoneal injection of dexmedetomidine 25 µg/kg. WB results showed that mBDNF, pTrkB/TrkB, and CREB were significantly decreased in the hippocampus of the sevoflurane group, which are significantly upregulated in the dexmedetomidine group. In the sevoflurane group, proBDNF, P75NRT, and RhoA were significantly increased, which were significantly lower than those in the dexmedetomidine group than those in the sevoflurane group. The expression BDNF was downregulated in the sevoflurane group, while the proBDNF was upregulated in the sevoflurane group. In the Morris water maze test, the escape latency of the sevoflurane group was significantly prolonged. In sevoflurane groups, the number of crossing platform was significantly reduced, the synaptic protein decreased significantly, and this effect was reversed in rats of the dexmedetomidine group. Dexmedetomidine could reduce synaptic plasticity decline in developing rats induced by sevoflurane, through downregulating the proBDNF-p75NTR-RhoA pathway and upregulating BDNF-TrkB-CREB.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dexmedetomidine/pharmacology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, trkB/metabolism , Receptors, Growth Factor/metabolism , Sevoflurane/toxicity , rhoA GTP-Binding Protein/metabolism , Animals , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Neurochem Int ; 139: 104789, 2020 10.
Article in English | MEDLINE | ID: mdl-32650025

ABSTRACT

Exposure to general anesthesia in early childhood may lead to adverse effects on adolescent neurocognition. This study investigated the effects of multiple inhalations of sevoflurane on long-term learning and memory in developing rats, and explored the mechanistic role of the tissue plasminogen activator (tPA)/plasminogen activator inhibitor-1 (PAI-1) fibrinolysis system and its regulatory relationship with the brain derived neurotrophic factor (BDNF) by activation of tropomysin related kinase B (TrkB). After rats were inhaled with sevoflurane for 2 h/d for three days, the expression levels of tPA, PAI-1, BDNF, its precursor(proBDNF), TrkB and phosphorylation of TrkB (p-TrkB) were detected at different time points. After 28 d, Morris water maze was used to examine learning and memory function; Golgi staining was used to investigate synaptic plasticity and synaptic-related proteins, such as Synapsin I(SYN1), growth associated protein 43(GAP-43), and postsynaptic density protein 95(PSD-95). Rats were given exogenous tPA and an inhibitor of PAI-1, TM5275. The results showed multiple inhalation of sevoflurane led to learning and memory dysfunction, downregulated the expression of the synaptic-related proteins, decreased dendritic spine density in the hippocampus, increased the expression level of proBDNF and PAI-1, and reduced expression of BDNF, tPA, and p-TrkB. Interestingly, tPA or TM5275 partially reversed the learning and memory dysfunction and the reduction of synaptic plasticity induced by sevoflurane exposure. Furthermore, they blocked the upregulation of proBDNF and PAI-1 protein expression and increased the expression of BDNF, tPA, and p-TrkB. The protective effect of tPA or TM5275 on rats following multiple sevoflurane inhalation was blocked by a TrkB inhibitor. Multiple inhalation of sevoflurane in rats inhibited the cleavage of proBDNF by disrupting the balance of the tPA/PAI-1 fibrinolysis system. This blocked the activation of the downstream TrkB signaling pathway and reduced hippocampal synaptic plasticity, leading to long-term learning and memory dysfunction. Therefore, Sevoflurane exposure could lead to learning and memory dysfunction by inhibiting BDNF cleavage via breaking the balance of tPA/PAI-1.


Subject(s)
Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Sevoflurane/toxicity , Tissue Plasminogen Activator/metabolism , Anesthetics, Inhalation/toxicity , Animals , Male , Maze Learning/physiology , Rats , Rats, Sprague-Dawley
6.
Brain Res Bull ; 161: 136-146, 2020 08.
Article in English | MEDLINE | ID: mdl-32433937

ABSTRACT

Excitotoxicity is one of the main mechanisms related to hypoxia/reoxygenation (H/R) injury. Excitatory amino acid transporter (EAAT)2 mainly distributes on astrocytes and plays an important role on glutamate reuptake and glutamate homeostasis. Midazolam has a neuroprotective effect in some neuropathological conditions. The present study aimed to detect the role of EAAT2 in the neuroprotective effect of midazolam in neonatal rat brain subjected to H/R. Pretreatment with midazolam reversed H/R-induced apoptosis and downregulation of EAAT2 mRNA and protein expression in the hippocampus. Pretreatment with dihydrokainic acid (a selective inhibitor of EAAT2) exacerbated apoptosis, and thus inhibited the neuroprotective effect of midazolam against H/R injury. We demonstrated for the first time that dysregulation of EAAT2 expression may be related to the neural injury induced by H/R in rat pups, and pretreatment with midazolam attenuated apoptosis and improved learning and memory partly due to regulating EAAT2 expression.


Subject(s)
Brain Injuries/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Hypoxia, Brain/metabolism , Midazolam/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Animals, Newborn , Brain Injuries/prevention & control , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Hypnotics and Sedatives/administration & dosage , Hypoxia, Brain/prevention & control , Rats , Rats, Sprague-Dawley
7.
Life Sci ; 242: 117151, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31843526

ABSTRACT

AIMS: Anaesthesia-related neurotoxicity in the developing brain is a controversial issue that has recently attracted much attention. Hemin plays a protective role in hypoxic and ischemic brain damage; however, its effects on sevoflurane-induced neurotoxicity remain unclear. Our aim was to investigate the mechanisms of sevoflurane neurotoxicity and potential neuroprotective roles of hemin upon sevoflurane exposure. MAIN METHODS: Hippocampi were harvested 18 h after sevoflurane exposure. Haem oxygenase 1 (HMOX1), superoxide dismutase 2 (SOD2), discs large MAGUK scaffold protein 4 (DLG4), phosphorylated Akt, Akt, cleaved caspase 3, and neuroglobin were detected by western blotting. A water maze test was used to assess learning and memory ability in P30 rats. KEY FINDINGS: Sevoflurane inhalation increased cleaved caspase 3 levels. Hemin treatment enhanced the antioxidant defence response, protecting rats from oxidative stress injury. Hemin plays its neuroprotective role via phosphoinositide 3-kinase (PI3K)/Akt signalling. A single inhalation of sevoflurane did not affect DLG4 expression, while hemin treatment did. Platform crossing increased in rats treated with hemin as well, which may be related to increased DLG4. Neuroglobin expression was not affected, suggesting that it may act upstream of PI3K/Akt signalling. SIGNIFICANCE: Our study demonstrates that hemin plays a protective role in anaesthesia-induced neurotoxicity by both inhibiting apoptosis via the PI3K/Akt pathway and increasing the expression of antioxidant enzymes, reducing oxidative damage. The results provide mechanistic insight into the effects of sevoflurane anaesthesia on the developing brain and suggest that hemin could help avoid these effects.


Subject(s)
Anesthetics, Inhalation/toxicity , Brain/drug effects , Hemin/pharmacology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sevoflurane/toxicity , Signal Transduction/drug effects , Administration, Inhalation , Animals , Animals, Newborn , Blotting, Western , Caspase 3/metabolism , Disks Large Homolog 4 Protein/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/enzymology , Maze Learning/drug effects , Neuroglobin/metabolism , Rats , Rats, Sprague-Dawley , Sevoflurane/antagonists & inhibitors , Superoxide Dismutase/metabolism
8.
Front Neurosci ; 13: 537, 2019.
Article in English | MEDLINE | ID: mdl-31191229

ABSTRACT

BACKGROUND: Many studies have reported that sevoflurane can increase neuronal apoptosis and result in cognitive deficits in rodents. Although neurotoxicity may be associated with mitochondrial dysfunction and oxidative stress, the exact mechanism remains unclear. In order to evaluate potential treatment therapies, we studied the effects of hemin on neurotoxicity of neonatal rat sevoflurane exposure. METHODS: Postnatal day (P) seven rats were assigned randomly to four groups; (1) group C: non-anesthesia, (2) group H: intraperitoneal hemin (50 mg kg-1) treatment on days 5 and 6, (3) group S: 3% sevoflurane exposure for 4 h, and (4) group SH: hemin treatment + sevoflurane exposure. The expression of neuroglobin in neonatal hippocampus was determined by western blot and immunohistochemistry. Neuroglobin was localized by immunofluorescence. Western blot for the expression of cleaved caspase-3 and TUNEL were used to detect neonatal hippocampal apoptosis, and cytochrome c was used to evaluate mitochondrial function. Drp-1 and Mfn-2 immunoblotting were used to assess mitochondrial dynamics. The Morris water maze test was performed to detect cognitive function in the rats on P30. RESULTS: Exposure to sevoflurane increased the expression of cleaved caspase-3, cytochrome c, and Drp1 in the neonatal hippocampus and resulted in cognitive deficiency but decreased expression of Mfn2. Hemin reduced apoptosis, improved mitochondrial dynamics and ameliorated the cognitive impairment caused by sevoflurane exposure. CONCLUSION: Hemin reduced neuronal apoptosis, improved mitochondrial dynamics and protected against cognitive deficits induced by sevoflurane in neonatal rats. This neuroprotective effect may be achieved by increasing the expression of neuroglobin.

9.
Drug Des Devel Ther ; 13: 153-160, 2019.
Article in English | MEDLINE | ID: mdl-30613136

ABSTRACT

BACKGROUND: Considerable evidences support the finding that the anesthesia reagent isoflurane increases neuronal cell death in young rats. Recent studies have shown that dexmedetomidine can reduce isoflurane-induced neuronal injury, but the mechanism remains unclear. We investigated whether isoflurane cause neurotoxicity to the central nervous system by regulating the N-methyl-D-aspartate receptor (NMDAR) and excitatory amino acid transporter1 (EAAT1) in young rats. Furthermore, we examined if dexmedetomidine could decrease isoflurane-induced neurotoxicity. METHODS: Neonatal rats (postnatal day 7, n=144) were randomly divided into four groups of 36 animals each: control (saline injection without isoflurane); isoflurane (2% for 4 h); isoflurane + single dose of dexmedetomidine (75 µg/kg, 20 min before the start of 2% isoflurane for 4 h); and isoflurane + dual doses of dexmedetomidine (25 µg/kg, 20 min before and 2 h after start of isoflurane at 2% for 4 h). Six neonates from each group were euthanatized at 2 h, 12 h, 24 h, 3 days, 7 days and 28 days post-anesthesia. Hippocampi were collected and processed for protein extraction. Expression levels of the NMDAR subunits NR2A and NR2B, EAAT1 and caspase-3 were measured by western blot analysis. RESULTS: Protein levels of NR2A, EAAT1 and caspase-3 were significantly increased in hippocampus of the isoflurane group from 2 h to 3 days, while NR2B levels were decreased. However, the -induced increase in NR2A, EAAT1 and caspase-3 and the decrease in NR2B in isoflurane-exposed rats were ameliorated in the rats treated with single or dual doses of dexmedetomidine. Isoflurane-induced neuronal damage in neonatal rats is due in part to the increase in NR2A and EAAT1 and the decrease in NR2B in the hippocampus. CONCLUSION: Dexmedetomidine protects the brain against the use of isoflurane through the regulation of NR2A, NR2B and EAAT1. However, using the same amount of dexmedetomidine, the trend of protection is basically the same.


Subject(s)
Dexmedetomidine/pharmacology , Glutamic Acid/metabolism , Isoflurane/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Dexmedetomidine/administration & dosage , Injections, Intraperitoneal , Isoflurane/pharmacology , Male , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
10.
Front Neurosci ; 12: 964, 2018.
Article in English | MEDLINE | ID: mdl-30618586

ABSTRACT

Numerous studies have demonstrated that general anesthetics might damage the nervous system, thus, the effect of general anesthetics on the developing brain has attracted much attention. Dexmedetomidine (Dex) exhibits a certain neuroprotective effect, but the mechanism is obscure. In our study, pregnant rats on gestational day 20 (G20) were exposed to 3% sevoflurane for 2 h or 4 h, and the neuronal apoptosis in hippocampal CA1 region of the offspring rats was detected by quantification of TUNEL positive cells and cleaved-caspase3 (cl-caspase3). Different doses of Dex were intraperitoneally injected before sevoflurane anesthesia; then, the expression of apoptotic-related proteins including BCL-2, BAX and cl-caspase3 as well as amyloid precursor protein (APP, a marker of axonal injury), p-CRMP-2 and CRMP-2 were measured at postnatal days 0, 1and 3 (P0, P1, and P3, respectively). As an antagonist of the bone morphgenetic proteins (BMP) receptor, DMH1 was co-administered with sevoflurane plus Dex to investigate whether BMP/SMAD is associated with the neuroprotective effects of Dex. The results showed that prenatal sevoflurane anesthesia for 4 h activated apoptosis transiently, as manifested by the caspase3 activity peaked on P1 and disappeared on P3. In addition, the expressions of APP and p-CRMP-2/CRMP-2 in postnatal rat hippocampus were significantly increased, which revealed that prenatal sevoflurane anesthesia caused axonal injury of offspring. The long-term learning and memory ability of offspring rats was also impaired after prenatal sevoflurane anesthesia. These damaging effects of sevoflurane could be mitigated by Dex and DMH1 reversed the neuroprotective effect of Dex. Our results indicated that prenatal exposure to 3% sevoflurane for 4 h increased apoptosis and axonal injury, even caused long-term learning and memory dysfunction in the offspring rats. Dex dose-dependently reduced sevoflurane- anesthesia-induced the neurotoxicity by activating the BMP/SMAD signaling pathway.

SELECTION OF CITATIONS
SEARCH DETAIL
...