Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Stroke Cerebrovasc Dis ; 32(12): 107395, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37839303

ABSTRACT

BACKGROUND: Our previous study found that hyperbaric oxygen (HBO) attenuated cognitive impairment in mice induced by cerebral ischemia-reperfusion injury (CIRI). However, its mechanism of action is not fully understood. In this study, we aimed to establish a rat model of cerebral ischemia-reperfusion, explore the possible role of ferroptosis in the pathogenesis of CIRI, and observe the effect of HBO on ferroptosis-mediated CIRI. METHODS: Sprague Dawley (SD) rats were randomly divided into control, model, Ferrostatin-1 (Fer-1), HBO and Fer-1+ HBO groups. Morris water maze, myelin basic protein (MBP) and ß-tubulin immunoreactivity were assessed to evaluate the neuroprotective effects of HBO on cerebral ischemia reperfusion injury. Ferroptosis were examined to investigate the mechanism underlying the effects of HBO. RESULTS: Our result showed that Fer-1 and HBO improved learning and memory ability in the navigation trail and probe trail of the Morris water maze and increased MBP and ß-tubulin immunoreactivity of the cortex in the model rats. The levels of ferritin, malondialdehyde (MDA) and glutathione (GSH) in the serum were also reversed by Fer-1 and HBO treatment. Mitochondrial cristae dissolution and vacuolization were observed in the model group by transmission electron microscopy and these conditions were improved in the Fer-1 and HBO groups. Furthermore, Fer-1 and HBO treatment reversed Prostaglandin-Endoperoxide Synthase 2 (PTGS2), Iron Responsive Element Binding Protein 2 (IREB2), acyl-CoA synthetase long chain family member 4 (ACSL4) and Solute Carrier Family 7 Member 11 (SLC7A11) mRNA levels and Transferrin Receptor 1 (TFR1), ferritin light chain (FTL), ferritin heavy chain 1 (FTH1), glutathione peroxidase 4 (GPX4), Nuclear factor E2-related factor 2 (Nrf2), lysophosphatidylcholine acyltransferase 3 (LPCAT3), c-Jun N-terminal kinase (JNK), phosphorylated c-Jun N-terminal kinase (P-JNK) phosphorylated Extracellular signal-regulated protein kinase (P-ERK) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) protein levels. The above changes were more pronounced in Fer-1+ HBOGroup. DISCUSSION: The results of the present study indicated that HBO improves cerebral ischemia-reperfusion injury in rats, which may be related to inhibition of ferroptosis. This also means that ferroptosis may become a new target of HBO against CIRI.


Subject(s)
Brain Ischemia , Ferroptosis , Hyperbaric Oxygenation , Reperfusion Injury , Rats , Mice , Animals , Rats, Sprague-Dawley , Hyperbaric Oxygenation/methods , Tubulin , Oxygen , Brain Ischemia/therapy , Extracellular Signal-Regulated MAP Kinases , JNK Mitogen-Activated Protein Kinases , Reperfusion Injury/pathology , 1-Acylglycerophosphocholine O-Acyltransferase
2.
PLoS One ; 17(11): e0276083, 2022.
Article in English | MEDLINE | ID: mdl-36355759

ABSTRACT

This study was to investigate the protective effect of hyperbaric oxygen (HBO) on HT22 and PC12 cell damage caused by oxygen-glucose deprivation/reperfusion-induced ferroptosis. A 2-h oxygen-glucose deprivation and 24-h reperfusion model on HT22 and PC12 cells was used to simulate cerebral ischemia-reperfusion injury. Cell viabilities were detected by Cell Counting Kit-8 (CCK-8) method. The levels of reactive oxygen species (ROS) and lipid reactive oxygen species (Lipid ROS) were detected by fluorescent probes Dihydroethidium (DHE) and C11 BODIPY 581/591. Iron Colorimetric Assay Kit, malondialdehyde (MDA) and glutathione (GSH) activity assay kits were used to detect intracellular iron ion, MDA and GSHcontent. Cell ferroptosis-related ultrastructures were visualized using transmission electron microscopy (TEM). Furthermore, PCR and Western blot analyses were used to detect the expressions of ferroptosis-related genes and proteins. After receiving oxygen-glucose deprivation/reperfusion, the viabilities of HT22 and PC12 cells were significantly decreased; ROS, Lipid ROS, iron ions and MDA accumulation occurred in the cells; GSH contents decreased; TEM showed that cells were ruptured and blebbed, mitochondria atrophied and became smaller, mitochondrial ridges were reduced or even disappeared, and apoptotic bodies appeared. And the expressions of Nrf2, SLC7A11 and GPX4 genes were reduced; the expressions of p-Nrf2/Nrf2, xCT and GPX4 proteins were reduced. Notably, these parameters were significantly reversed by HBO, indicating that HBO can protect HT22 cells and PC12 cells from damage caused by oxygen-glucosedeprivation/reperfusion via the inhibition of Nrf2/System Xc-/GPX4 axis-mediated ferroptosis.


Subject(s)
Ferroptosis , Hyperbaric Oxygenation , Rats , Animals , PC12 Cells , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Oxygen/metabolism , Glucose , Phospholipid Hydroperoxide Glutathione Peroxidase , Reperfusion , Iron/metabolism , Lipids
3.
Mol Med Rep ; 22(5): 4254-4264, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32901878

ABSTRACT

Our previous study demonstrated that hyperbaric oxygen (HBO) improves heart function predominantly through reducing oxygen stress, modulating energy metabolism and inhibiting cell apoptosis. The present study aimed to investigate the protective effects of HBO on mitochondrial function and autophagy using rats with a ligated left anterior descending artery. The cardioprotective effects of HBO were mainly evaluated using ELISA, fluorescent probes, transmission electron microscopy and reverse transcription­quantitative PCR (RT­qPCR). HBO pretreatment for 14 days (once a day) using a 0.25 MPa chamber improved mitochondrial morphology and decreased the number of autophagic vesicles, as observed using a transmission electron microscope. HBO pretreatment significantly increased the levels of ATP, ADP, energy charge and the opening of the mitochondrial permeability transition pore, but decreased the levels of AMP, cytochrome c and reactive oxygen species. Moreover, HBO pretreatment significantly increased the gene or protein expression levels of eIF4E­binding protein 1, mammalian target of rapamycin (mTOR), mitochondrial DNA, NADH dehydrogenase subunit 1, mitofusin 1 and mitofusin 2, whereas it decreased the gene or protein expression levels of autophagy­related 5 (Atg5), cytochrome c, dynamin­related protein 1 and p53, as determined using RT­qPCR or immunohistochemistry. In conclusion, HBO treatment was observed to protect cardiomyocytes during myocardial ischemia­reperfusion injury (MIRI) by preventing mitochondrial dysfunction and inhibiting autophagy. Thus, these results provide novel evidence to support the use of HBO as a potential agent for the mitigation of MIRI.


Subject(s)
Hyperbaric Oxygenation/methods , Mitochondria/metabolism , Myocardial Reperfusion Injury/therapy , Animals , Autophagy , Disease Models, Animal , Energy Metabolism , Gene Expression Regulation , Male , Myocardial Reperfusion Injury/metabolism , Rats , Treatment Outcome
4.
Life Sci ; 241: 117170, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31838137

ABSTRACT

AIMS: In this study, we investigate the effect and underlying mechanism of hyperbaric oxygen (HBO) treatment on a model of repeated cerebral ischemia-reperfusion injury (IR). MAIN METHODS: Eighty rats were randomly separated into sham, vehicle, hyperbaric air (HBA; 0.25 MPa, 60 min), and HBO (0.25 MPa, 60 min) groups. Repeated cerebral IR was induced by ligating the right and left bilateral common carotid arteries for 10 min and then allowing reperfusion for 10 min. This pattern was repeated three times. The neuroprotective effects of HBO were assessed by animal behavior, neuron morphology, inflammatory markers, intracellular calcium ion content, and autophagy-related protein and gene expression. KEY FINDINGS: Our result showed that HBO improved learning and memory in the navigation trail and probe trail of the Morris water maze, and these findings were supported by the observation data from 2,3,5-Triphenyltet-razolium chloride staining, Nissl staining, and electron microscopic. Importantly, we found that HBO reduced excessive autophagy in the prefrontal cortex, which was evidenced by activating of the mammalian target of the rapamycin (mTOR) and 4E-BP1, as well as suppression of LC3II and ATG5. Moreover, HBO significantly inhibited the cerebral IR-induced inflammatory reaction. Furthermore, HBO treatment modulated autophagy pathway-related factors, including producing a decrease in the intracellular calcium ion concentration and p53 level; meanwhile, the levels of BDNF and p-Akt were increased. SIGNIFICANCE: Our results indicated that HBO protected against IR-induced neuron injury by attenuating autophagy, inflammation, and calcium overload. These results provide a new mechanism and laboratory evidence for clinical treatment of VD.


Subject(s)
Autophagy , Brain Ischemia/complications , Cognitive Dysfunction/prevention & control , Disease Models, Animal , Hyperbaric Oxygenation/methods , Neuroprotective Agents , Reperfusion Injury/complications , Animals , Behavior, Animal , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Female , Male , Mice , Rats , Rats, Sprague-Dawley
5.
Med Chem ; 15(5): 459-471, 2019.
Article in English | MEDLINE | ID: mdl-30569869

ABSTRACT

Hyperbaric Oxygenation Therapy (HBOT) is used as an adjunctive method for multiple diseases. The method meets the routine treating and is non-invasive, as well as provides 100% pure oxygen (O2), which is at above-normal atmospheric pressure in a specialized chamber. It is well known that in the condition of O2 deficiency, it will induce a series of adverse events. In order to prevent the injury induced by anoxia, the capability of offering pressurized O2 by HBOT seems involuntary and significant. In recent years, HBOT displays particular therapeutic efficacy in some degree, and it is thought to be beneficial to the conditions of angiogenesis, tissue ischemia and hypoxia, nerve system disease, diabetic complications, malignancies, Carbon monoxide (CO) poisoning and chronic radiation-induced injury. Single and combination HBOT are both applied in previous studies, and the manuscript is to review the current applications and possible mechanisms of HBOT. The applicability and validity of HBOT for clinical treatment remain controversial, even though it is regarded as an adjunct to conventional medical treatment with many other clinical benefits. There also exists a negative side effect of accepting pressurized O2, such as oxidative stress injury, DNA damage, cellular metabolic, activating of coagulation, endothelial dysfunction, acute neurotoxicity and pulmonary toxicity. Then it is imperative to comprehensively consider the advantages and disadvantages of HBOT in order to obtain a satisfying therapeutic outcome.


Subject(s)
Hyperbaric Oxygenation , Animals , Cardiovascular Diseases/therapy , Humans , Hypoxia/therapy , Ischemia/therapy , Neovascularization, Physiologic/physiology , Nervous System Diseases/therapy
6.
Neurochem Res ; 43(3): 531-539, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29313212

ABSTRACT

In this study, we examined whether hyperbaric oxygen (HBO2) plays a detoxification role in withdrawal symptoms in a morphine-dependent rat model. The model was established through injections of morphine at increasing doses for 7 days. Withdrawal symptoms were induced by naloxone injection on the 8th day. The detoxification effect of HBO2 was evaluated using the withdrawal symptom scores, biochemical indices and neurotransmitters. Compared with the model group, HBO2 therapy significantly attenuated the withdrawal symptom scores, body weight loss and the level of norepinephrine level, whereas it increased the dopamine level and tyrosine hydroxylase expression in the nucleus accumbens. Moreover, HBO2 therapy substantially alleviated the NO, NOS, cAMP, and cGMP levels. Our findings indicate that HBO2 can effectively alleviate withdrawal symptoms induced by morphine dependence, and these effects may be attributed to the modulation of monoaminergic neurotransmitters and the suppression of the NO-cGMP signaling pathway.


Subject(s)
Hyperbaric Oxygenation , Morphine/pharmacology , Neurotransmitter Agents/metabolism , Nitric Oxide/metabolism , Nucleus Accumbens/drug effects , Animals , Male , Morphine Dependence/metabolism , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Substance Withdrawal Syndrome/drug therapy
7.
Neurochem Res ; 42(4): 1240-1253, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28078611

ABSTRACT

Our previous study demonstrated that hyperbaric oxygen (HBO) improved cognitive impairments mainly by regulating oxidative stress, inflammatory responses and aging-related gene expression. However, a method for preventing cognitive dysfunction has yet to be developed. In the present study, we explored the protective effects of HBO on the cholinergic system and apoptosis in D-galactose (D-gal)-treated mice. A model of aging was established via systemic intraperitoneal injection of D-gal daily for 8 weeks. HBO was administered during the last 2 weeks of D-gal injection. Our results showed that HBO in D-gal-treated mice significantly improved behavioral performance on the open field test and passive avoidance task. Studies on the potential mechanisms of this effect showed that HBO significantly reduced oxidative stress and blocked the nuclear factor-κB pathway. Moreover, HBO significantly increased the levels of choline acetyltransferase and acetylcholine and decreased the activity of acetylcholinesterase in the hippocampus. Furthermore, HBO markedly increased expression of the anti-apoptosis protein Bcl-2 and glial fibrillary acidic protein meanwhile decreased expression of the pro-apoptosis proteins Bax and caspase-3. Importantly, there was a significant reduction in expression of Aß-related genes, such as amyloid precursor protein, ß-site amyloid cleaving enzyme-1 and cathepsin B mRNA. These decreases were accompanied by significant increases in expression of neprilysin and insulin-degrading enzyme mRNA. Moreover, compared with the Vitamin E group, HBO combined with Vitamin E exhibited significant difference in part of the above mention parameters. These findings suggest that HBO may act as a neuroprotective agent in preventing cognitive impairments.


Subject(s)
Apoptosis/physiology , Cholinergic Neurons/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/prevention & control , Galactose/toxicity , Hyperbaric Oxygenation/methods , Animals , Apoptosis/drug effects , Cholinergic Neurons/drug effects , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Random Allocation , Vitamin A/pharmacology , Vitamin A/therapeutic use
8.
Cancer Lett ; 381(2): 331-40, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27523281

ABSTRACT

Dysregulation in iron metabolism can lead to a wide range of diseases including cancer. Iron-regulatory proteins (IRPs) and iron responsive elements (IREs) have been established as post-transcriptional regulators of intracellular iron homeostasis. The roles of other pathways involved in this process, however, remain largely unknown. Here we report that epidermal growth factor receptor (EGFR), an oncogenic driver, binds to and regulates the subcellular distribution of transferrin receptor 1(TfR1) through its tyrosine kinase activity and thus is required for cellular iron import. Inactivation of EGFR reduces the cell surface TfR1 expression, which leads to decreased iron import due to impaired TfR1-mediated iron uptake. This damaged iron assimilation results in cell cycle arrest and growth inhibition, which can be partially rescued by non-Tf-bound iron supplements. Evaluation of non-small cell lung cancer samples reveals a positive correlation between EGFR activation and membrane TfR1 expression. Our findings uncover a new role of EGFR in modulating cellular iron homeostasis through redistribution of TfR1, which is essential for cancer development and progression.


Subject(s)
Adenocarcinoma/enzymology , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Proliferation , ErbB Receptors/metabolism , Iron/metabolism , Lung Neoplasms/enzymology , Receptors, Transferrin/metabolism , A549 Cells , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antigens, CD/genetics , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Cytoplasm/enzymology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Homeostasis , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Receptors, Transferrin/genetics , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
9.
Neurosci Lett ; 600: 220-5, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26101828

ABSTRACT

Parkinson's disease (PD) is a common movement disorder in the elderly. In the present study, we examined whether the combination of hyperbaric oxygen (HBO) and madopar therapy provided a neuroprotective effect on dopaminergic neurons in the substantia nigra using a rat model of PD. Rotational assessments revealed that both HBO and combination therapy significantly attenuated apomorphine-induced turning in PD rats. Our results indicated that the combination therapy increased glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) activities and reduced the malondialdehyde (MDA) content in the SN. Furthermore, the combination therapy resulted in significant protection against the loss of neurons, and specifically tyrosine hydroxylase (TH)-positive neurons, in the SN and also alleviated the production of glial fibrillary acidic protein (GFAP). The levels of Bcl-2 were increased and Bax were decreased following the HBO or combination therapy. In brief, the neuroprotective effect of combined therapy with HBO and madopar against 6-OHDA-induced PD rats may rely on its ability to reduce oxidative stress and protect against Bax/Bcl-2-mediated apoptosis.


Subject(s)
Benserazide/therapeutic use , Hyperbaric Oxygenation , Levodopa/therapeutic use , Neuroprotective Agents/therapeutic use , Oxidopamine/therapeutic use , Parkinson Disease/therapy , Animals , Apomorphine/pharmacology , Combined Modality Therapy , Drug Combinations , Glial Fibrillary Acidic Protein/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxidation , Male , Malondialdehyde/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Oxidative Stress , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Wistar , Stereotyped Behavior , Substantia Nigra/metabolism , Substantia Nigra/pathology , Superoxide Dismutase/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL