Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neuroimmune Pharmacol ; 18(3): 366-382, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37318680

ABSTRACT

Ischemic stroke is a cerebrovascular lesion caused by local ischemia and hypoxia. Diabetes mellitus (DM) is a chronic inflammatory disease that disturbs immune homeostasis and predisposes patients to ischemic stroke. The mechanism by which DM exacerbates stroke remains unclear, although it may involve disturbances in immune homeostasis. Regulatory T cells (Tregs) play a regulatory role in many diseases, but the mechanism of Tregs in diabetes complicated by stroke remains unclear. Sodium butyrate is a short-chain fatty acid that increases Treg levels. This study examined the role of sodium butyrate in the prognosis of neurological function in diabetic stroke and the mechanism by which Tregs are amplified in the bilateral cerebral hemispheres. We evaluated the brain infarct volume, observed 48-h neuronal injury and 28-day behavioral changes, and calculated the 28-day survival rate in mice. We also measured Treg levels in peripheral blood and brain tissue, recorded changes in the blood‒brain barrier and water channel proteins and neurotrophic changes in mice, measured cytokine levels and peripheral B-cell distribution in bilateral hemispheres and peripheral blood, and examined the polarization of microglia and the distribution of peripheral T-cell subpopulations in bilateral hemispheres. Diabetes significantly exacerbated the poor prognosis and neurological deficits in mice with stroke, and sodium butyrate significantly improved infarct volume, prognosis, and neurological function and showed different mechanisms in brain tissue and peripheral blood. The potential regulatory mechanism in brain tissue involved modulating Tregs/TGF-ß/microglia to suppress neuroinflammation, while that in peripheral blood involved improving the systemic inflammatory response through Tregs/TGF-ß/T cells.

2.
J Neuroimmune Pharmacol ; 18(1-2): 90-99, 2023 06.
Article in English | MEDLINE | ID: mdl-35895245

ABSTRACT

Diabetes is an independent risk factor for stroke and amplifies inflammation. Diabetic stroke is associated with a higher risk of death and worse neural function. The identification of effective anti-inflammatory molecules with translational advantages is particularly important to promote perioperative neurorestorative effects. Applying molecular hydrogen, we measured blood glucose levels before and after middle cerebral artery occlusion (MCAO), 48-h cerebral oedema and infarct volumes, as well as 28-day weight, survival and neurological function. We also measured the levels of TLR4, NF-κB p65, phosphorylated NF-κB p65, catecholamines, acetylcholine and inflammatory factors. All measurements comprehensively showed the positive effect and translational advantage of molecular hydrogen on diabetic stroke. Molecular hydrogen improved the weight, survival and long-term neurological function of rats with diabetic stroke and alleviated changes in blood glucose levels before and after middle cerebral artery occlusion (MCAO), but no difference in circadian rhythm was observed. Molecular hydrogen inhibited the phosphorylation of NF-κB and significantly reduced inflammation. Molecular hydrogen mediates neurorestorative effects after stroke in diabetic rats. The effect is independent of circadian rhythms, indicating translational advantages. The molecular mechanism is related to the TLR4/NF-κB pathway and inflammation. Molecular hydrogen (H2) affects outcomes of ischemic stroke with diabetes mellitus (DM).


Subject(s)
Diabetes Mellitus, Experimental , Stroke , Rats , Animals , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Signal Transduction , Blood Glucose , Diabetes Mellitus, Experimental/complications , Rats, Sprague-Dawley , Stroke/drug therapy , Stroke/complications , Inflammation , Hydrogen/pharmacology
3.
Neural Regen Res ; 17(9): 1991-1997, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35142688

ABSTRACT

Diabetes mellitus is an independent risk factor for ischemic stroke. Both diabetes mellitus and stroke are linked to systemic inflammation that aggravates patient outcomes. Stellate ganglion block can effectively regulate the inflammatory response. Therefore, it is hypothesized that stellate ganglion block could be a potential therapy for ischemic stroke in diabetic subjects. In this study, we induced diabetes mellitus in rats by feeding them a high-fat diet for 4 successive weeks. The left middle cerebral artery was occluded to establish models of ischemic stroke in diabetic rats. Subsequently, we performed left stellate ganglion block with 1% lidocaine using the percutaneous posterior approach 15 minutes before reperfusion and again 20 and 44 hours after reperfusion. Our results showed that stellate ganglion block did not decrease the blood glucose level in diabetic rats with diabetes mellitus but did reduce the cerebral infarct volume and the cerebral water content. It also improved the recovery of neurological function, increased 28-day survival rate, inhibited Toll like receptor 4/nuclear factor kappa B signaling pathway and reduced inflammatory response in the plasma of rats. However, injection of Toll like receptor 4 agonist lipopolysaccharide 5 minutes before stellate ganglion block inhibited the effect of stellate ganglion block, whereas injection of Toll like receptor 4 inhibitor TAK242 had no such effect. We also found that stellate ganglion block performed at night had no positive effect on diabetic ischemic stroke. These findings suggest that stellate ganglion block is a potential therapy for diabetic ischemic stroke and that it may be mediated through the Toll like receptor 4/nuclear factor kappa B signaling pathway. We also found that the therapeutic effect of stellate ganglion block is affected by circadian rhythm.

4.
Eur J Pharmacol ; 914: 174664, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34883075

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI)-induced acute lung injury (ALI) is a critical condition, and inflammation and apoptosis play essential roles. Molecular hydrogen (H2) exerts anti-inflammatory and anti-apoptotic effects. Our previous work has shown that 42% H2 can improve TBI. In the current study, we tested the hypothesis that inhalation of hydrogen (42% H2, 21% O2, balanced nitrogen) for 1 h per day can improve TBI-induced ALI. METHODS: Sprague-Dawley male rats were randomly divided into 3 groups. Except for the sham group (group S), rats were subjected to a fluid percussion injury (FPI) and the H2 treatment group were given inhaled hydrogen for 1 h per day. We evaluated the lung function, pyroptosis and apoptosis at 24 h, 48 h and 72 h. RESULTS: Compared with group S, the rats in the TBI group (group T) showed obvious pulmonary edema after a TBI. Inhalation of high-concentration hydrogen significantly improved the rats. During this process, rats had some tendency to heal on their own, and H2 also accelerated the self-healing process. Lung injury scores, oxygenation index and pulmonary edema were consistent. Compared with group S, the pyroptosis-related proteins Caspase-1, apoptosis-associated speck-like protein containing CARD (ASC) and Gasdermin-D (GSDM-D) in the lung tissues of the rats in group T were significantly increased after a TBI. In the H2 treatment group (group H), these proteins were significantly decreased. The levels of IL-1ß and IL-18 were significantly increased after TBI while in group H were significantly decreased. At the same time, cleaved caspase-3 and BCL-2/Bax were also changed after H2 treatment. These demonstrates the powerful ameliorating effect of H2 on pyroptosis, apoptosis and systemic inflammation. However, rats also had tendency to heal on their own, and H2 also accelerated the self-healing process at the same time. CONCLUSIONS: H2 improves TBI-ALI, and the mechanism may be due to the decrease of both pyroptosis and apoptosis and the alleviation of inflammation. These findings provide a reference and evidence for the use of H2 in TBI-ALI patients in the intensive care unit (ICU).


Subject(s)
Acute Lung Injury , Brain Injuries, Traumatic/complications , Hydrogen , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Administration, Inhalation , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Hydrogen/administration & dosage , Hydrogen/pharmacology , Interleukin-1beta/metabolism , Nitrogen/administration & dosage , Oxygen/administration & dosage , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pulmonary Edema/etiology , Pulmonary Edema/therapy , Pyroptosis/drug effects , Rats , Rats, Sprague-Dawley , Treatment Outcome
5.
Neural Regen Res ; 16(8): 1574-1581, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33433486

ABSTRACT

Studies have shown that hyperglycemia aggravates brain damage by affecting vascular endothelial function. However, the precise mechanism remains unclear. Male Sprague-Dawley rat models of diabetes were established by a high-fat diet combined with an intraperitoneal injection of streptozotocin. Rat models of traumatic brain injury were established using the fluid percussion method. Compared with traumatic brain injury rats without diabetic, diabetic rats with traumatic brain injury exhibited more severe brain injury, manifested as increased brain water content and blood-brain barrier permeability, the upregulation of heme oxygenase-1, myeloperoxidase, and Bax, the downregulation of occludin, zona-occludens 1, and Bcl-2 in the penumbra, and reduced modified neurological severity scores. The intraperitoneal injection of a nitric oxide synthase inhibitor N(5)-(1-iminoethyl)-L-ornithine (10 mg/kg) 15 minutes before brain injury aggravated the injury. These findings suggested that nitric oxide synthase plays an important role in the maintenance of cerebral microcirculation, including anti-inflammatory, anti-oxidative stress, and anti-apoptotic activities in diabetic rats with traumatic brain injury. The experimental protocols were approved by the Institutional Animal Care Committee of Harbin Medical University, China (approval No. ky2017-126) on March 6, 2017.

6.
Brain Res ; 1730: 146651, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31926128

ABSTRACT

Reactive oxygen species, inflammation, and apoptosis are major contributors to secondary injuries that follow traumatic brain injury (TBI) in diabetic patients. Hydrogen (H2) can selectively neutralize reactive oxygen species and downregulate inflammatory and apoptotic factors. Therefore, we investigated the effects of inhaled high and low concentrations of hydrogen on neurological function after TBI in diabetic rats and the potential mechanism. We found that the inhalation of high concentrations of H2 significantly improved outcomes following TBI in diabetic rats. The inhalation of 42% H2 for one hour per day for 48 h significantly reduced brain edema, decreased the extravasation of sodium fluorescein, and reduced oxidative stress markers (p < 0.05). In addition, the inhalation of a high concentration of H2 (42% for one hour per day for 7 days) improved neurological deficits (p < 0.05) and reduced the expression of apoptotic protein markers (p < 0.05). However, the inhalation of 3% H2 did not yield significant effects. These results showed that the inhalation of 42% H2 can alleviate nerve damage and improve neurological function after TBI in diabetic rats. Therefore, the inhalation of a high concentration of H2 may be associated with the treatment of traumatic brain injuries.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries, Traumatic/psychology , Brain/drug effects , Diabetes Complications/psychology , Hydrogen/administration & dosage , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain/pathology , Brain Edema/prevention & control , Brain Injuries, Traumatic/complications , Male , Neurons/drug effects , Rats, Sprague-Dawley
7.
Med Gas Res ; 9(2): 80-87, 2019.
Article in English | MEDLINE | ID: mdl-31249256

ABSTRACT

Central nervous system injuries are a leading cause of death and disability worldwide. Although the exact pathophysiological mechanisms of various brain injuries vary, central nervous system injuries often result in an inflammatory response, and subsequently lead to brain damage. This suggests that neuroprotection may be necessany in the treatment of multiple disease models. The use of medical gases as neuroprotective agents has gained great attention in the medical field. Medical gases include common gases, such as oxygen, hydrogen and carbon dioxide; hydrogen sulphide and nitric oxide that have been considered toxic; volatile anesthetic gases, such as isoflurane and sevoflurane; and inert gases like helium, argon, and xenon. The neuroprotection from these medical gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Nevertheless, the transition into the clinical practice is still lagging. This delay could be attributed to the contradictory paradigms and the conflicting results that have been obtained from experimental models, as well as the presence of inconsistent reports regarding their safety. In this review, we summarize the potential mechanisms underlying the neuroprotective effects of medical gases and discuss possible candidates that could improve the outcomes of brain injury.


Subject(s)
Brain Injuries/drug therapy , Gases/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Gases/chemistry , Helium/chemistry , Helium/therapeutic use , Humans , Hydrogen/chemistry , Hydrogen/therapeutic use , Hyperbaric Oxygenation , Isoflurane/chemistry , Isoflurane/therapeutic use , Neuroprotective Agents/chemistry
8.
J Neuroinflammation ; 16(1): 36, 2019 Feb 13.
Article in English | MEDLINE | ID: mdl-30760300

ABSTRACT

BACKGROUND: This study investigated whether therapeutic hypercapnia (TH) ameliorated blood-brain barrier (BBB) damage and improved the neurologic outcome in a rat model of lateral fluid percussion injury (FPI), and explored the possible underlying mechanism. METHODS: Rats underwent lateral FPI and received inhalation of 30%O2-70%N2 or 30%O2-N2 plus CO2 to maintain arterial blood CO2 tension (PaCO2) between 80 and 100 mmHg for 3 h. To further explore the possible mechanisms for the protective effects of TH, a PKC inhibitor staurosporine or PKCαß inhibitor GÖ6976 was administered via intracerebral ventricular injection. RESULTS: TH significantly improved neurological function 24 h, 48 h, 7 d, and 14 d after FPI. The wet/dry ratio, computed tomography values, Evans blue content, and histological lesion volume were significantly reduced by TH. Moreover, numbers of survived neurons and the expression of tight junction proteins (ZO-1, occludin, and claudin-5) were significantly elevated after TH treatment at 48-h post-FPI. TH significantly increased the expression of protein kinase Cε (PKCε) at 48-h post-FPI, but did not significantly change the expression of PKCα and PKCßII. PKC inhibitor staurosporine (but not the selective PKCαß inhibitor-GÖ6976) inhibited the protective effect of TH. CONCLUSIONS: Therapeutic hypercapnia is a promising candidate that should be further evaluated for clinical treatment. It not only protects the traumatic penumbra from secondary injury and improves histological structure but also maintains the integrity of BBB and reduces neurologic deficits after trauma in a rat model of FPI.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , Carbon Dioxide/therapeutic use , Hypercapnia , Protein Kinase C-epsilon/metabolism , Animals , Blood Pressure/drug effects , Brain Edema/etiology , Brain Edema/therapy , Brain Injuries, Traumatic/diagnostic imaging , Carbazoles/therapeutic use , Disease Models, Animal , Heart Rate/drug effects , Male , Neurologic Examination , Oxygen/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Staurosporine/therapeutic use , Time Factors , Tomography Scanners, X-Ray Computed
9.
World Neurosurg ; 119: e765-e773, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30096509

ABSTRACT

OBJECTIVE: Traumatic brain injury (TBI) is a devastating neurologic injury and remains a major cause of death in the world. Secondary injury after TBI is associated with long-term disability in patients with TBI. This study evaluated adrenomedullin (AM) on secondary injury and neurologic functional outcome in rats after TBI. METHODS: Forty-eight Sprague Dawley rats were randomly assigned into 3 groups: sham, TBI, and TBI with AM groups. TBI was induced by fluid percussion injury, and AM was intravenously injected. Neurologic function was examined at 2, 3, and 7 days after TBI. Enzyme-linked immunosorbent assay was used to test tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-8 levels in the brain. Brain edema and blood-brain barrier (BBB) permeability in brain tissue were tested. Western blot was used to examine the expression of aquaporin-4, phosphorylated myosin light-chain, and cleaved caspase-3. Terminal deoxynucleotidyl transferase dUTP nick end labeling was used to test the apoptosis. RESULTS: Compared with the sham group, TNF-α, IL-1ß, and IL-6 levels, brain edema, BBB permeability, neurologic examination scores, terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells, and expression of aquaporin-4, phosphorylated myosin light-chain, and cleaved caspase-3 significantly increased in the TBI group. AM treatment significantly inhibited TBI-induced effects. CONCLUSIONS: AM can improve neurologic function and ameliorate brain injury in rats with TBI. AM exerts its neuroprotective effect via its anti-inflammatory and antiapoptotic effect.


Subject(s)
Adrenomedullin/pharmacology , Brain Injuries, Traumatic/prevention & control , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain Diseases/physiopathology , Brain Edema/prevention & control , Neurologic Examination , Nociception/physiology , Posture/physiology , Rats, Sprague-Dawley , Reaction Time/physiology , Walking/physiology
10.
Brain Res ; 1644: 9-14, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27163721

ABSTRACT

Cerebral ischemia/reperfusion (I/R) injury could cause neural apoptosis that involved the signaling cascades. Cytochrome c release from the mitochondria and the followed activation of caspase 9 and caspase 3 are the important steps. Now, a new mitochondrial protein, apoptosis-inducing factor (AIF), has been shown to have relationship with the caspase-independent apoptotic pathway. In this study, we investigated the protective effects of propofol through inhibiting AIF-mediated apoptosis induced by whole cerebral I/R injury in rats. 120 Wistar rats that obtained the permission of the animal care committee of Harbin Medical University were randomly divided into three groups: sham group (S group), cerebral ischemia/reperfusion injury group (I/R group), and propofol treatment group (P group). Propofol (1.0mg/kg/min) was administered intravenously for 1h before the induction of ischemia in P group. The apoptotic rate in three groups was detected by flow cytometry after 24h of reperfusion. The mitochondrial membrane potential (MMP) changes were detected via microplate reader. The expressions of B-cell leukemia-2 (Bcl-2), Bcl-2 associated X protein (Bax) and AIF were evaluated using Western blot after 6h, 24h and 48h of reperfusion. The results of our study showed that apoptotic level was lower in P group compared with I/R group and propofol could protect MMP. The ratio of Bcl-2/Bax was significantly higher in P group compared with I/R group. The translocation of AIF from mitochondrial to nucleus was lower in P group than that in I/R group. Our findings suggested that the protective effects of propofol on cerebral I/R injury might be associated with inhibiting translocation of AIF from mitochondrial to the nucleus in hippocampal neurons.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Brain Ischemia/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Propofol/administration & dosage , Animals , Brain Ischemia/prevention & control , Hippocampus/drug effects , Hippocampus/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
11.
Brain Res ; 1622: 1-6, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26100336

ABSTRACT

Post-operative cerebral edema is a threat for patients performed gliomas resection. Some studies have shown that general anesthesia drugs, such as, propofol had neuroprotective effect. Aquaporin-4 (AQP4) and Aquaporin-9 (AQP9) play an important role in maintaining brain water homeostasis under various conditions. The aim of this study was to compare the effect of propofol or sevoflurane on expression of AQP4 and AQP9 in patients performed gliomas resection. 30 patients performed gliomas resection were included in this study. The patients were randomly divided into two groups: propofol group and sevoflurane group. Fresh human gliomas specimens were obtained and hematoxylin eosin (HE) staining, immunohistochemical staining and Western blot analysis were used for observation of the expression of AQP4 and AQP9. The immunohistochemical staining of the sections showed that the percentage of AQP4 positive cells in the propofol group (14.3±4.61%) was significantly lower than that in sevoflurane group (37.3±10.01%) (n=15, P<0.05). There was no significant difference in the percentage of AQP9 positive cells in propofol group and sevoflurane group (25.8±2.67 versus 28.1±7.81%, n=15, P>0.05). Western blot analysis confirmed the immunohistochemistry results. AQP4 protein level in propofol group was significantly lower than that in sevoflurane group (1.4±0.13 versus 1.7±0.12, P<0.05). Western blot analysis did not show any difference of expression of AQP9 protein between the propofol group and sevoflurane group (2.0±0.13 versus 2.1±0.13, P>0.05, n=6). AQP4 expression was lower in patients of propofol group than that in sevoflurane group. Our results suggested that propofol could inhibit the expression of AQP4.


Subject(s)
Brain Neoplasms/surgery , Brain/drug effects , Glioma/surgery , Methyl Ethers/therapeutic use , Neuroprotective Agents/therapeutic use , Propofol/therapeutic use , Aquaporin 4/metabolism , Aquaporins/metabolism , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/surgery , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/prevention & control , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Glioma/metabolism , Glioma/pathology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Immunohistochemistry , Male , Middle Aged , Random Allocation , Sevoflurane
12.
Mol Med Rep ; 12(2): 2181-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25892548

ABSTRACT

α-Calcitonin gene-related peptide (α-CGRP) is a 37 amino-acid neuropeptide that is primarily released from C-type sensory neurons. α-CGRP exerts multiple modulatory effects on immune responses and visceral organ function, but the role of exogenous α-CGRP in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has remained to be elucidated. Forty-eight rats were randomized to receive continuous intraperitoneal infusion of α-CGRP (0.4 µg/kg/min) or normal saline for 30 min, followed by intratracheal injection of 0.5 mg/kg LPS or saline. There were four groups of animals: The saline-saline (S-S) group; the saline-α-CGRP (S-C) group; the LPS-saline (L-S) group and the LPS-α-CGRP (L-C) group. Mean arterial pressure and arterial blood gases were assessed prior to α-CGRP and LPS administration and every hour following LPS treatment. After 4 h, bronchoalveolar lavage was performed and used to assess total cell count and levels of tumor necrosis factor-α, interleukin-1ß, intracellular cell adhesion molecule 1 and macrophage inflammatory protein 2. Lung tissue was also collected for assessing wet-to-dry (W/D) ratio, histology and Evans blue (EB) dye extravasation. Pulmonary α-CGRP concentration and α-CGRP receptor expression were also examined, and inducible cyclic adenosine monophosphate early repressor (ICER) and TNF-α mRNA expression levels were measured. Treatment with exogenous α-CGRP improved oxygenation during LPS-induced ALI. Correspondingly, histological injury, total cell count, inflammatory cytokine levels, W/D ratio and EB dye extravasation were also significantly reduced. α-CGRP receptor 1 expression was noted in pulmonary endothelial cells and alveolar macrophages and α-CGRP receptor expression levels were decreased during ALI, whereas pulmonary α-CGRP expression was continuously increased. Furthermore, exogenous α-CGRP induced upregulation of ICER during LPS-induced ALI. In conclusion, exogenous α-CGRP improved oxygenation and ameliorated lung damage in LPS-induced ALI, and these effects were associated with the upregulation of ICER.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/immunology , Calcitonin Gene-Related Peptide/therapeutic use , Immunologic Factors/therapeutic use , Lipopolysaccharides/immunology , Lung/drug effects , Lung/immunology , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Calcitonin Gene-Related Peptide/immunology , Cyclic AMP Response Element Modulator/genetics , Gene Expression Regulation/drug effects , Immunologic Factors/immunology , Interleukin-1beta/analysis , Interleukin-1beta/immunology , Lung/metabolism , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Calcitonin Gene-Related Peptide/genetics , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
13.
Brain Res ; 1594: 108-14, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25451088

ABSTRACT

Propofol is a commonly used intravenous anesthetic that has been demonstrated to be neuroprotective against cerebral ischemia-reperfusion (I/R) injury. It remains unclear whether this protective effect has any relationship with the prevention of neuronal mitochondrial deoxyribonucleic acid (mtDNA) deletion. In this study, 81 Wistar rats were randomly divided into three groups (n = 27 each): sham (S group), ischemia/reperfusion (I/R group), or propofol (P group). Cerebral ischemia was induced by clamping the bilateral common carotid arteries for 10 min. A polymerase chain reaction (PCR) was conducted to determine mtDNA deletion. The mitochondrial membrane potential (MMP) changes were detected via microplate reader. The neuronal ultrastructure was visualized via electron microscope. MMP significantly decreased after I/R (P<0.05 compared with the S group). Severe damage to the ultrastructure of neuronal mitochondria was observed in cerebral I/R injury. When propofol (1.0mg/kg/min) was administered intravenously for 1h prior to the induction of I/R, the neuronal structure and MMP were well preserved, and mtDNA deletion was reduced after ischemia/reperfusion injury compared with the I/R group (P<0.05). These data suggested that propofol prevented mtDNA deletion and preserved a normal structure and MMP, which are important for normal mitochondrial function and increase neuronal resistance to I/R injury.


Subject(s)
Brain Ischemia/pathology , DNA, Mitochondrial/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Propofol/pharmacology , Reperfusion Injury/pathology , Animals , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Neurons/ultrastructure , Polymerase Chain Reaction , Rats , Rats, Wistar
14.
Lab Invest ; 93(12): 1339-49, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24126891

ABSTRACT

Hypercapnic acidosis may attenuate ventilator-induced lung oxidative stress injury and alveolar cell apoptosis, but the underlying mechanisms are poorly understood. We examined the effects of hypercapnic acidosis on the role of apoptosis signal-regulating kinase 1 (ASK1), which activates the c-Jun N-terminal kinase (JNK) and p38 cascade in both apoptosis and oxidative reactions, in high-pressure ventilation stimulated rat lungs. Rats were ventilated with a peak inspiratory pressure (PIP) of 30 cmH2O for 4 h and randomly given FiCO2 to achieve normocapnia (PaCO2 at 35-45 mm Hg) or hypercapnia (PaCO2 at 80-100 mm Hg); normally ventilated rats with PIP of 15 cmH2O were used as controls. Lung injury was quantified by gas exchange, microvascular leaks, histology, levels of inflammatory cytokines, and pulmonary oxidative reactions. Apoptosis through the ASK1-JNK/p38 mitogen-activated protein kinase (MAPK) cascade in type II alveolar epithelial cells (AECIIs) were evaluated by examination of caspase-3 activation. The results showed that injurious ventilation caused significant lung injury, including deteriorative oxygenation, changes of histology, and the release of inflammatory cytokines. In addition, the high-pressure mechanical stretch also induced apoptosis and caspase-3 activation in the AECIIs. Hypercapnia attenuated these responses, suppressing the ASK1 signal pathways with its downstream kinase phosphorylation of p38 MAPK and JNK, and caspase-3 activation. Thus, hypercapnia can attenuate cell apoptosis and oxidative stress damage in rat lungs during injurious ventilation, at least in part, due to the suppression of the ASK1-JNK/p38 MAPK pathways.


Subject(s)
Hypercapnia , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/prevention & control , Acidosis/metabolism , Acidosis/pathology , Animals , Apoptosis/physiology , Hemodynamics , Hypercapnia/diagnosis , Hypercapnia/metabolism , Hypercapnia/pathology , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Stress, Mechanical , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...