Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Phytomedicine ; 130: 155733, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38759314

ABSTRACT

BACKGROUND: The proinflammatory response induced by macrophages plays a crucial role in the development of sepsis and the resulting multiorgan dysfunction. Identifying new regulatory targets for macrophage homeostasis and devising effective treatment strategies remains a significant challenge in contemporary research. PURPOSE: This study aims to identify new regulatory targets for macrophage homeostasis and develop effective strategies for treating sepsis. STUDY DESIGN AND METHODS: Macrophage infiltration in septic patients and in lungs, kidneys, and brains of caecum ligation and puncture (CLP)-induced septic mice was observed using CIBERSORT and immunofluorescence (IF). Upon integrating the MSigDB database and GSE65682 dataset, differently expressed macrophage-associated genes (DEMAGs) were identified. Critical DEMAGs were confirmed through machine learning. The protein level of the critical DEMAG was detected in PBMCs of septic patients, RAW264.7 cells, and mice lungs, kidneys, and brains using ELISA, western blot, immunohistochemistry, and IF. siRNA was applied to investigate the effect of the critical DEMAG in RAW264.7 cells. A natural product library was screened to find a compound targeting the critical DEMAG protein. The binding of compounds and proteins was analyzed through molecular docking, molecular dynamics simulations, CETSA, and MST analysis. The therapeutic efficacy of the compounds against sepsis was then evaluated through in vitro and in vivo experiments. RESULTS: Macrophage infiltration was inversely correlated with survival in septic patients. The critical differentially expressed molecule RasGRP1 was frequently observed in the PBMCs of septic patients, LPS-induced RAW264.7 cells, and the lungs, kidneys, and brains of septic mice. Silencing RasGRP1 alleviated proinflammatory response and oxidative stress in LPS-treated RAW264.7 cells. Catechin Hydrate (CH) was identified as an inhibitor of RasGRP1, capable of maintaining macrophage homeostasis and mitigating lung, kidney, and brain damage during sepsis. CONCLUSION: This study demonstrates that RasGRP1, a novel activator of macrophage proinflammatory responses, plays a crucial role in the excessive inflammation and oxidative stress associated with sepsis. CH shows potential for treating sepsis by inhibiting RasGRP1.


Subject(s)
Catechin , Guanine Nucleotide Exchange Factors , Macrophages , Sepsis , Animals , Sepsis/drug therapy , Mice , Humans , RAW 264.7 Cells , Macrophages/drug effects , Macrophages/metabolism , Male , Guanine Nucleotide Exchange Factors/metabolism , Catechin/pharmacology , Multiple Organ Failure/drug therapy , Molecular Docking Simulation , Kidney/drug effects , Mice, Inbred C57BL , Disease Models, Animal , Lung/drug effects
2.
Heliyon ; 10(9): e30172, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38707378

ABSTRACT

Background: Sepsis-associated acute lung injury (ALI) and acute kidney injury (AKI) are common complications that significantly impact patient prognosis. Danlou tablet (DLT) is a traditional herbal preparation with anti-inflammatory and antioxidant properties. However, its therapeutic potential in sepsis remains unknown. Methods: The impact of DLT on ALI and AKI was evaluated using the cecal ligation and puncture (CLP) experimental sepsis animal model. The effects of DLT on macrophages were observed through LPS-stimulated RAW264.7 cell line. Inflammatory cytokines, oxidative stress indicators, HE, PAS, and DHE staining, lung wet-to-dry weight ratio, and serum creatinine and urea nitrogen levels were used to assess tissue injury. Network pharmacology, molecular docking, and molecular dynamics simulations were used to explore the potential regulatory mechanisms of DLT in sepsis. Western blot and immunohistochemical staining were used to validate the expression of mechanism-related proteins. Results: DLT inhibited the inflammatory response and oxidative stress, improved structural and functional abnormalities in lung and kidney tissues in CLP mice, and alleviated pro-inflammatory responses of LPS-stimulated macrophages. PARP1 and HMGB1 were identified as key regulatory targets. The results of in vitro and in vivo experiments suggest that DLT can effectively inhibit PARP1/HMGB1 and improve sepsis-associated ALI and AKI. Conclusion: The present study demonstrated that DLT suppressed pro-inflammatory responses of macrophage and alleviated ALI and AKI in the CLP mice by inhibiting the transition activation of PARP1/HMGB1. These findings partially elucidate the mechanism of DLT in sepsis-associated ALI and AKI and further clarify the active components of DLT, thereby providing a scientific theoretical basis for treating sepsis with DLT.

3.
J Ethnopharmacol ; 322: 117610, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38122915

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The QiShengYiQi pill (QSYQ) is a traditional Chinese medicinal formulation. The effectiveness and safety of QSYQ in treating respiratory system disorders have been confirmed. Its pharmacological actions include anti-inflammation, antioxidative stress, and improving energy metabolism. However, the mechanism of QSYQ in treating sepsis-induced acute lung injury (si-ALI) remains unclear. AIM OF THE STUDY: Si-ALI presents a clinical challenge with high incidence and mortality rates. This study aims to confirm the efficacy of QSYQ in si-ALI and to explore the potential mechanisms, providing a scientific foundation for its application and insights for optimizing treatment strategies and identifying potential active components. MATERIALS AND METHODS: The impact of QSYQ on si-ALI was evaluated using the cecal ligation and puncture (CLP) experimental sepsis animal model. The effects of QSYQ on endothelial cells were observed through coculturing with LPS-stimulated macrophage-conditioned medium. Inflammatory cytokine levels, HE staining, Evans blue staining, lung wet/dry ratio, and cell count and protein content in bronchoalveolar lavage fluid were used to assess the degree of lung injury. Network pharmacology was utilized to investigate the potential mechanisms of QSYQ in treating si-ALI. Western blot and immunofluorescence analyses were used to evaluate barrier integrity and validate mechanistically relevant proteins. RESULTS: QSYQ reduced the inflammation and alleviated pulmonary vascular barrier damage in CLP mice (all P < 0.05). A total of 127 potential targets through which QSYQ regulates si-ALI were identified, predominantly enriched in the RAGE pathway. The results of protein-protein interaction analysis suggest that COX2, a well-established critical marker of ferroptosis, is among the key targets. In vitro and in vivo studies demonstrated that QSYQ mitigated ferroptosis and vascular barrier damage in sepsis (all P < 0.05), accompanied by a reduction in oxidative stress and the inhibition of the COX2 and RAGE (all P < 0.05). CONCLUSIONS: This study demonstrated that QSYQ maintains pulmonary vascular barrier integrity by inhibiting ferroptosis in CLP mice. These findings partially elucidate the mechanism of QSYQ in si-ALI and further clarify the active components of QSYQ, thereby providing a scientific theoretical basis for treating si-ALI with QSYQ.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Ferroptosis , Sepsis , Mice , Animals , Endothelial Cells/metabolism , Cyclooxygenase 2/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Lung , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Lipopolysaccharides/pharmacology
4.
Front Pharmacol ; 13: 940261, 2022.
Article in English | MEDLINE | ID: mdl-36188533

ABSTRACT

Background: Ferroptosis has a vital role in sepsis, but the mechanism is not known. Understanding the mechanism of ferroptosis during sepsis will aid in developing improved therapeutic strategies. Methods: We used the Gene Expression Omnibus database and FerrDb database to obtain ferroptosis-related differentially expressed genes (DEGs) between sepsis patients and healthy volunteers (HVs). Analyses of PPI networks, functional enrichment, as well as use of the MCODE algorithm were used to identify key ferroptosis-related DEGs. Expression of key ferroptosis-related DEGs was verified using: GSE57065 and GSE65682 datasets; rats in which ferroptosis was induced with erastin; sepsis-induced acute lung injury (siALI) rats. The effects of acupoint catgut embedding (ACE) on ferroptosis and expression of key ferroptosis-related DEGs in the lungs of siALI rats were also observed. A Cox proportional hazard model was used to verify the effect of key ferroptosis-related DEGs on the survival of sepsis patients. Cytoscape was used to construct ceRNA networks and gene-transcription factor networks. Results: Between sepsis patients and HVs, we identified 33 ferroptosis-related DEGs. According to analyses of PPI networks and the MCODE algorithm, we obtained four modules, of which the most significant module contained nine ferroptosis-related DEGs. Functional-enrichment analyses showed that four of the nine DEGs were enriched in the MAPK signaling pathway: MAPK14, VEGFA, TGFBR1, and DUSP1. We verified expression of these four genes in GSE57065 and GSE65682 datasets and ferroptosis rats. In addition, expression of these four genes and that of the oxidative-stress indicators GSSG and MDA was upregulated, and glutathione peroxidase-4 (GPX4) expression was downregulated, in siALI rats, but ACE reversed these changes. The Cox proportional hazard model showed that survival of sepsis patients in the high-risk group was shorter than that in the low-risk group. We found that the XIST-hsa-let-7b-5p-TGFBR1/DUSP1 ceRNA network and transcription factor E2F1 may be important regulators of these four DEGs. Conclusion: Our results suggest that MAPK14, VEGFA, TGFBR1, and DUSP1 may be key regulatory targets of ferroptosis in sepsis, and that ACE pretreatment may be antioxidant treatment for sepsis and alleviate ferroptosis. These findings provide a basis for further ferroptosis-related study in sepsis and provide new targets for its treatment.

5.
Respir Res ; 22(1): 258, 2021 Oct 03.
Article in English | MEDLINE | ID: mdl-34602075

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is a key contributing factor to poor survival in lung transplantation recipients. Mitochondrial dysfunction is recognized as a critical mediator in the pathogenesis of diabetic lung ischemia-reperfusion (IR) injury. The protective effects of adiponectin have been demonstrated in our previous study, but the underlying mechanism remains unclear. Here we demonstrated an important role of mitophagy in the protective effect of adiponectin during diabetic lung IR injury. METHODS: High-fat diet-fed streptozotocin-induced type 2 diabetic rats were exposed to adiponectin with or without administration of the SIRT1 inhibitor EX527 following lung transplantation. To determine the mechanisms underlying the action of adiponectin, rat pulmonary microvascular endothelial cells were transfected with SIRT1 small-interfering RNA or PINK1 small-interfering RNA and then subjected to in vitro diabetic lung IR injury. RESULTS: Mitophagy was impaired in diabetic lungs subjected to IR injury, which was accompanied by increased oxidative stress, inflammation, apoptosis, and mitochondrial dysfunction. Adiponectin induced mitophagy and attenuated subsequent diabetic lung IR injury by improving lung functional recovery, suppressing oxidative damage, diminishing inflammation, decreasing cell apoptosis, and preserving mitochondrial function. However, either administration of 3-methyladenine (3-MA), an autophagy antagonist or knockdown of PINK1 reduced the protective action of adiponectin. Furthermore, we demonstrated that APN affected PINK1 stabilization via the SIRT1 signaling pathway, and knockdown of SIRT1 suppressed PINK1 expression and compromised the protective effect of adiponectin. CONCLUSION: These data demonstrated that adiponectin attenuated reperfusion-induced oxidative stress, inflammation, apoptosis and mitochondrial dysfunction via activation of SIRT1- PINK1 signaling-mediated mitophagy in diabetic lung IR injury.


Subject(s)
Adiponectin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Lung Injury/prevention & control , Lung Transplantation/adverse effects , Lung/drug effects , Mitochondria/drug effects , Mitophagy/drug effects , Reperfusion Injury/prevention & control , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/enzymology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Lung/enzymology , Lung/pathology , Lung Injury/enzymology , Lung Injury/etiology , Lung Injury/pathology , Male , Mitochondria/enzymology , Mitochondria/pathology , Oxidative Stress/drug effects , Protein Kinases/genetics , Protein Kinases/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism
6.
Surgery ; 165(5): 1014-1026, 2019 05.
Article in English | MEDLINE | ID: mdl-30824287

ABSTRACT

BACKGROUND: Lung ischemia-reperfusion injury is a complex pathophysiologic process associated with high morbidity and mortality. We have demonstrated elsewhere that diabetes mellitus aggravated ischemia-induced lung injury. Oxidative stress and mitochondrial dysfunction are drivers of diabetic lung ischemia-reperfusion injury; however, the pathways that mediate these events are unexplored. In this study using a high-fat diet-fed model of streptozotocin-induced type 2 diabetes in rats, we determined the effect of hydrogen sulfide on lung ischemia-reperfusion injury with a focus on Sirtuin3 signaling. METHODS: Rats with type 2 diabetes were exposed to GYY4137, a slow release donor of hydrogen sulfide with or without administration of the Sirtuin3 short hairpin ribonucleic acid plasmid, and then subjected to a surgical model of ischemia-reperfusion injury of the lung (n = 8). Lung function, oxidative stress, inflammation, cell apoptosis, and mitochondrial function were measured. RESULTS: Compared with nondiabetic rats, animals with type 2 diabetes at baseline exhibited significantly decreased Sirtuin3 signaling in lung tissue and increased oxidative stress, apoptosis, inflammation, and mitochondrial dysfunction (P < .05 each). In addition, further impairment in Sirtuin3 signaling was found in diabetic rats subjected to this model of lung ischemia-reperfusion. Simultaneously, the indexes showed further aggravation. Treatment with hydrogen sulfide restored Sirtuin3 expression and decreased lung ischemia-reperfusion injury in animals with type 2 diabetes mellitus by improving lung functional recovery, decreasing oxidative damage, suppressing inflammation, ameliorating cell apoptosis, and preserving mitochondrial function (P < .05). Conversely, these protective effects were largely reversed in Sirtuin3 knockdown rats. CONCLUSION: Impaired lung Sirtuin3 signaling associated with type 2 diabetic conditions was further attenuated by an ischemia-reperfusion insult. Hydrogen sulfide ameliorated reperfusion-induced oxidative stress and mitochondrial dysfunction via activation of Sirtuin3 signaling, thereby decreasing lung ischemia-reperfusion damage in rats with a model of type II diabetes.


Subject(s)
Diabetes Mellitus, Type 2/complications , Hydrogen Sulfide/pharmacology , Lung Injury/prevention & control , Mitochondria/drug effects , Sirtuins/metabolism , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Drug Evaluation, Preclinical , Humans , Hydrogen Sulfide/therapeutic use , Lung/drug effects , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , Mitochondria/metabolism , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , RNA, Small Interfering/metabolism , Rats , Reperfusion Injury/complications , Signal Transduction/drug effects , Sirtuins/genetics , Streptozocin/toxicity
7.
Int Immunopharmacol ; 69: 95-102, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30690345

ABSTRACT

Previous studies have shown that ghrelin, a peptide produced in the stomach, attenuates acute lung injury (ALI) in various animal models, and that some of these effects are associated with inhibition of the nuclear factor κB signaling pathway. This study investigated whether ghrelin exerts beneficial effects on hemorrhagic shock (HS)-induced ALI by modulating nuclear factor κB inhibitor kinase/nuclear factor κB inhibitor/nuclear factor κB (IKK/IκBα/NF-κB) pathway activity. HS was induced in male SD rats by withdrawing blood to a mean arterial pressure (MAP) of 40 mm Hg for 1 h; rats then received ghrelin (10 nmol/kg) or vehicle intravenously and were resuscitated with the shed blood and an equal volume of Ringer lactate solution followed by observation for 2 h. After resuscitation, samples were collected and analyzed for lung histopathology, wet to dry weight ratio (W/D), bronchoalveolar lavage fluid (BALF) protein, neutrophil infiltration, plasma inflammatory cytokines (TNF-α and IL-6), and cytoplasmic phosphorylated IKKß, IκBα, phosphorylated IκBα and nuclear NF-κB expression. Compared to those in the two sham groups, lung injury, W/D, BALF protein, neutrophil infiltration, plasma TNF-α and IL-6 levels, and IKK/IκBα/NF-κB pathway activation were significantly increased in HS rats. After ghrelin administration, all parameters analyzed were decreased compared to those without ghrelin in HS rats. Moreover, ghrelin alleviated the decreased MAP after resuscitation compared to that in HS rats. Exogenous ghrelin attenuates the inflammatory response and acute lung injury after HS. These beneficial effects appear to be mediated through inhibition of IKK/IκBα/NF-κB signaling.


Subject(s)
Acute Lung Injury/drug therapy , Ghrelin/therapeutic use , I-kappa B Kinase/metabolism , Lung/drug effects , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Shock, Hemorrhagic/drug therapy , Administration, Intravenous , Animals , Disease Models, Animal , Humans , Interleukin-6/blood , Lung/pathology , Male , Neutrophil Infiltration , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/blood
8.
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
9.
Neurocrit Care ; 24(3): 459-71, 2016 06.
Article in English | MEDLINE | ID: mdl-26561305

ABSTRACT

INTRODUCTION: Propofol exhibits neuroprotective effects mediated by the inhibition of excitatory amino acid (EAA) neurotransmitter release and potentiation of inhibitory amino acid (IAA) neurotransmitters. To our knowledge, this is the first study to investigate the effects of propofol on the EAA and IAA balance in neurogenic pulmonary edema (NPE). METHODS: Sixty male Wistar rats were randomized to Sham, NPE, Low-dose propofol, and High-dose propofol groups. NPE was induced via rapid injection of autologous blood (0.5 ml) into the cisterna magna. The Low- and High-dose propofol groups were pretreated with boluses of 2 and 5 mg kg(-1), respectively, prior to blood injection, followed by continuous propofol infusion at 6 and 15 mg kg(-1) h(-1), respectively. The mean arterial pressure (MAP), heart rate, intracranial pressure (ICP), peak inspiratory pressure (PIP), and arterial blood gases were continuously recorded. After 2 h, the lung wet-to-dry weight ratio, total protein concentration in the bronchoalveolar lavage fluid (BALF), brain water content, cortical EAA and IAA levels, chest X-ray, and histological staining of lung sections were evaluated. RESULTS: Blood injections into the cisterna magna induced NPE and hemodynamic changes. Propofol alleviated the increases in the MAP, ICP, and PIP, improved oxygenation and histopathological changes, ameliorated pulmonary and cerebral edema, increased the IAA brain levels, and decreased the ratio of Glu to γ-aminobutyric acid. CONCLUSIONS: The current findings suggest that propofol improves NPE likely via IAA accumulation and the regulation of EAA and IAA balance, which may represent an effective treatment for NPE.


Subject(s)
Brain Edema/drug therapy , Brain/drug effects , Brain/metabolism , Neuroprotective Agents/pharmacology , Neurotransmitter Agents/metabolism , Propofol/pharmacology , Pulmonary Edema/drug therapy , Subarachnoid Hemorrhage/complications , Animals , Brain Edema/etiology , Disease Models, Animal , Excitatory Amino Acids/antagonists & inhibitors , Glutamic Acid/drug effects , Male , Neuroprotective Agents/administration & dosage , Neurotransmitter Agents/agonists , Neurotransmitter Agents/antagonists & inhibitors , Propofol/administration & dosage , Pulmonary Edema/etiology , Rats , Rats, Wistar , gamma-Aminobutyric Acid/drug effects
10.
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
11.
BMC Anesthesiol ; 15: 67, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25924944

ABSTRACT

BACKGROUND: We have proved that hypercapnic acidosis (a PaCO2 of 80-100 mmHg) protects against ventilator-induced lung injury in rats. However, there remains uncertainty regarding the appropriate target PaCO2 or if greater CO2 "doses" (PaCO2 > 100 mmHg) demonstrate this effect. We wished to determine whether severe acute hypercapnic acidosis can reduce stretch-induced injury, as well as the role of nuclear factor-κB (NF-κB) in the effects of acute hypercapnic acidosis. METHODS: Fifty-four rats were ventilated for 4 hours with a pressure-controlled ventilation mode set at a peak inspiratory pressure (PIP) of 30 cmH2O. A gas mixture of carbon dioxide with oxygen (FiCO2 = 4-5%, FiCO2 = 11-12% or FiCO2 = 16-17%; FiO2 = 0.7; balance N2) was immediately administered to maintain the target PaCO2 in the NC (a PaCO2 of 35-45 mmHg), MHA (a PaCO2 of 80-100 mmHg) and SHA (a PaCO2 of 130-150 mmHg) groups. Nine normal or non-ventilated rats served as controls. The hemodynamics, gas exchange and inflammatory parameters were measured. The role of NF-κB pathway in hypercapnic acidosis-mediated protection from high-pressure stretch injury was then determined. RESULTS: In the NC group, high-pressure ventilation resulted in a decrease in PaO2/FiO2 from 415.6 (37.1) mmHg to 179.1 (23.5) mmHg (p < 0.001), but improved by MHA (379.9 ± 34.5 mmHg) and SHA (298.6 ± 35.3 mmHg). The lung injury score in the SHA group (7.8 ± 1.6) was lower than the NC group (11.8 ± 2.3, P < 0.05) but was higher than the MHA group (4.4 ± 1.3, P < 0.05). Compared with the NC group, after 4 h of high pressure ventilation, the MHA and SHA groups had decreases in MPO activity of 67% and 33%, respectively, and also declined the levels of TNF-α (58% versus 72%) and MIP-2 (76% versus 60%) in the BALF. Additionally, both hypercapnic acidosis groups reduced stretch-induced NF-κB activation (p < 0.05) and significantly decreased lung ICAM-1 expression (p < 0.05). CONCLUSIONS: Moderate hypercapnic acidosis (PaCO2 maintained at 80-100 mmHg) has a greater protective effect on high-pressure ventilation-induced inflammatory injury. The potential mechanisms may involve alterations in NF-κB activity.


Subject(s)
Acidosis, Respiratory/metabolism , Hypercapnia/metabolism , NF-kappa B/metabolism , Ventilator-Induced Lung Injury/prevention & control , Acidosis, Respiratory/physiopathology , Animals , Carbon Dioxide/metabolism , Disease Models, Animal , Hypercapnia/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Pulmonary Gas Exchange/physiology , Rats , Rats, Wistar , Severity of Illness Index , Tumor Necrosis Factor-alpha/metabolism
12.
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
15.
Med Hypotheses ; 80(2): 158-61, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23207183

ABSTRACT

Neurogenic pulmonary edema (NPE) is an acute and serious complication after a central nervous system insult with high mortality. The pronounced activation of sympathetic nervous system and the release of vasoactive substances are necessary prerequisites for the development of NPE. We introduce a hypothesis that stellate ganglion block (SGB) may prevent NPE development on the basis of the inhibition of sympathetic overactivation, reduction of the concentration of norepinephrine and attenuation of baroreflex sensitivity, and improve the outcome by improving cerebral blood flow and pulmonary circulation and maintaining cardiovascular stability. In clinical practice, the guidance technique and close monitoring might guarantee the safety of SGB. If our hypothesis is supported by further experiments, this may open a new doorway for the treatment of NPE.


Subject(s)
Pulmonary Edema/drug therapy , Pulmonary Edema/prevention & control , Stellate Ganglion/drug effects , Sympatholytics/pharmacology , Cerebrovascular Circulation/drug effects , Humans , Models, Biological , Norepinephrine/metabolism , Pulmonary Circulation/drug effects , Pulmonary Edema/physiopathology
16.
Cell Physiol Biochem ; 28(2): 315-22, 2011.
Article in English | MEDLINE | ID: mdl-21865739

ABSTRACT

BACKGROUND: Previous studies demonstrate that macrophages synthesis and release catecholamines, which regulate the immune responses in an autocrine manner. These responses are mediated in part by ß-adrenoceptors expressed on macrophages. Some ß-adrenoceptor antagonists are commonly used in clinical conditions. Here we investigated whether the chronic administration of ß-adrenoceptor antagonists upregulate adrenergic system of alveolar macrophage and the potential mechanims. METHODS: Propranolol (30 mg/kg·d) or atenolol (5 mg/kg·d) was administered by gavage to rats for 4 weeks. Then alveolar macrophages were isolated and the expression of ß(1) or ß(2)-adrenoceptor was detected by flow cytometric analysis. Dopamine ß-hydroxylase expression was assessed by Western blot assay and the concentrations of noradrenaline, IL-6, and TNF-α in cell supernatants were measured using ELISA after 2 h or 24 h exposure of alveolar macrophages to 100 ng/ml lipopolysaccharide (LPS). RESULTS: Propranolol increased the mean fluorescence intensity (MFI) of ß(1), ß(2)-adrenoceptor and the frequency of ß(1)-,ß(2)- adrenoceptor positive macrophages. However, only the MFI of ß(1)-adrenoceptor and the frequency of ß(1)-adrenoceptor positive macrophages were increased by atenolol. Furthermore, both propranolol and atenolol promoted LPS-mediated dopamine ß-hydroxylase protein expression and increased noradrenaline production in rat alveolar macrophages. This was accompanied by increased LPS-mediated IL-6 and TNF-α production in cell supernatants of alveolar macrophages. CONCLUSION: These findings demonstrate that propranolol or atenolol upregulates alveolar macrophage adrenergic system, and the response may be ß(1)-adrenergic receptor subtype dependent.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Macrophages, Alveolar/drug effects , Receptors, Adrenergic, beta-1/metabolism , Up-Regulation/drug effects , Animals , Atenolol/pharmacology , Dopamine beta-Hydroxylase/metabolism , Fluorescent Dyes/chemistry , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophages, Alveolar/metabolism , Norepinephrine/metabolism , Propranolol/pharmacology , Rats , Rats, Wistar , Receptors, Adrenergic, beta-2/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Brain Res ; 1410: 24-32, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21783180

ABSTRACT

Propofol is an intravenous anesthetic with neuroprotective effects against cerebral ischemia-reperfusion (I/R) injury. Few studies regarding the neuroprotective and neurobehavioral effects of propofol have been conducted, and the underlying mechanisms are still unclear. Because I/R may result in neuronal apoptosis, the apoptosis regulatory genes B-cell leukemia-2 (Bcl-2) and Bcl-2-associated X protein (Bax) may be involved in the neuroprotective process. In this study, 120 Wistar rats were randomly divided into three groups (sham, I/R-induced, and propofol-treated). Cerebral ischemia was induced by clamping the bilateral common carotid arteries for 10min. Propofol (1.0mg/kg/min) was administered intravenously for 1h before the induction of ischemia. Neuronal damage was evaluated by neurobehavioral scores and histological examination of the brain sections at the level of the dorsal hippocampus at 6h, 24h, 48h, 72h, 4days, 5days, 6days, and 7days after I/R. The apoptotic rate of hippocampal neurons was detected by flow cytometry. The expression of Bcl-2 and Bax was evaluated using immunohistochemical and Western blot methods. The results of this study showed that neurobehavioral scores were higher in propofol-treated rats compared with I/R-induced rats with no propofol treatment. Moreover, the hippocampal expression of Bcl-2 was significantly higher, while the expression of Bax was significantly lower in propofol-treated rats compared with I/R-induced rats at 24h after ischemia. Hence, this study suggests that the neuroprotective effects of propofol against neuronal apoptosis may be a consequence of the regulation of Bcl-2 and Bax.


Subject(s)
Anesthetics, Intravenous/therapeutic use , Behavior, Animal/drug effects , Brain Ischemia/drug therapy , Propofol/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Reperfusion Injury/drug therapy , bcl-2-Associated X Protein/metabolism , Anesthetics, Intravenous/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/drug effects , Neurons/metabolism , Propofol/pharmacology , Rats , Rats, Wistar , Reperfusion Injury/metabolism
18.
Eur J Pharmacol ; 653(1-3): 16-20, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21114997

ABSTRACT

Propofol has been shown to exert neuroprotective effects. Delayed rectifier potassium current (I(K)) was reported to be closely related to neuronal damage. This study was designed to test the effects of propofol on I(K) in rat parietal cortical neurons and the involvement of PKC in this activity. Whole-cell patch-clamp recordings were performed in rat parietal cortical neurons. The amplitudes of I(K) were recorded before and after the addition of different concentrations of propofol. Propofol concentration-dependently inhibited I(K) with an IC50 value of 36.3±2.7 µM. Moreover, propofol caused a downward shift of the I-V curve of I(K) in a concentration dependent manner. The kinetics of I(K) was altered by propofol, with decreased activation and delayed recovery of I(K). Pretreatment with calphostin-C (a non-selective inhibitor of PKC) or PKC epsilon translocation inhibitor peptide (PKC epsilon inhibitor) abrogated the inhibition of I(K) by propofol. In conclusion, propofol inhibited I(K) via the activation of PKC epsilon in rat cerebral parietal cortical neurons.


Subject(s)
Delayed Rectifier Potassium Channels/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Propofol/pharmacology , Protein Kinase C-epsilon/drug effects , Animals , Dose-Response Relationship, Drug , Female , Male , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Parietal Lobe/drug effects , Parietal Lobe/metabolism , Patch-Clamp Techniques , Propofol/administration & dosage , Protein Kinase C-epsilon/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...