Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Transl Cancer Res ; 13(4): 1737-1761, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38737676

ABSTRACT

Background: The presence of portal vein tumor thrombus (PVTT) is a significant indicator of advanced-stage hepatocellular carcinoma (HCC). Unfortunately, the prediction of PVTT occurrence remains challenging, and there is a lack of comprehensive research exploring the underlying mechanisms of PVTT formation and its association with immune infiltration. Methods: Our approach involved analyzing single-cell sequencing data, applying high dimensional weighted gene co-expression network analysis (hdWGCNA), and identifying key genes associated with PVTT development. Furthermore, we constructed competing endogenous RNA (ceRNA) networks and employed weighted gene co-expression network analysis (WGCNA), as well as three machine-learning techniques, to identify the upstream regulatory microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) of the crucial mRNAs. We employed fuzzy clustering of time series gene expression data (Mfuzz), gene set variation analysis (GSVA), and cell communication analysis to uncover significant signaling pathways involved in the activation of these important mRNAs during PVTT development. In addition, we conducted immune infiltration analysis, survival typing, and drug sensitivity analysis using The Cancer Genome Atlas (TCGA) cohort to gain insights into the two patient groups under study. Results: Through the implementation of hdWGCNA, we identified 110 genes that was closely associated with PVTT. Among these genes, TMEM165 emerged as a crucial candidate, and we further investigated its significance using COX regression analysis. Furthermore, through machine learning techniques and survival analysis, we successfully identified the upstream regulatory miRNA (hsa-miR-148a) and lncRNA (LINC00909) that targeted TMEM165. These findings shed light on the complex regulatory network surrounding TMEM165 in the context of PVTT. Moreover, we conducted CIBERSORT analysis, which unveiled correlations between TMEM165 and immune infiltration in HCC patients. Specifically, TMEM165 exhibited associations with various immune cell populations, including memory B cells and CD8+ T cells. Additionally, we observed implications for immune function, particularly in relation to immune checkpoints, within the context of HCC. Conclusions: The regulatory axis involving TMEM165, hsa-miR-148a, and LINC00909 emerges as a crucial determinant in the development of PVTT in HCC patients, and it holds significant implications for prognosis. Furthermore, alterations in the TMEM165/hsa-miR-148a/LINC00909 regulatory axis exhibit a strong correlation with immune infiltration within the HCC tumor microenvironment, leading to immune dysfunction and potential failure of immunotherapy.

2.
J Am Heart Assoc ; 11(7): e023787, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35289183

ABSTRACT

Background Post-resuscitation syndrome, involves a severe inflammatory response following successful cardiopulmonary resuscitation. The potential mechanism of Vitamin C (VitC) after cardiopulmonary resuscitation on myocardial and cerebral function, duration of survival is undefined. Methods and Results A first set of experiments were done in 18 male Sprague-Dawley rats for the investigation of short-term follow-up, randomized into 3 groups: (1) sham; (2) controls; (3) VitC. Ventricular fibrillation was electrically induced and untreated for 6 minutes. Cardiopulmonary resuscitation including chest compression and mechanical ventilation were then initiated and continued for 8 minutes followed by defibrillation. At 5 minutes after return of spontaneous circulation, either VitC (200 mg/kg) or placebo was administered by intravenous infusion with a syringe pump for half an hour. There were significant improvements in myocardial function and buccal microcirculation in rats treated with VitC after return of spontaneous circulation 4 hours compared with controls. VitC inhibited proinflammatory cytokines (interleukin-6 and tumor necrosis factor-α), SDC-1 (Syndecan-1), and hyaluronic acid in plasma compared with controls (P<0.01). VitC decreased reactive oxygen species production and inhibited p38/MAPK (mitogen-activated protein kinase) pathway phosphorylation. A second set with 20 animals was used for assessing the neurological deficit score after return of spontaneous circulation 72 hours, randomized into 2 groups: 1) controls; 2) VitC. The survival rate and neurological deficit score after return of spontaneous circulation 72 hours were improved in VitC-treated animals compared with those of the control group. Conclusions VitC reduces the severity of post-resuscitation myocardial and cerebral dysfunction and improves the survival. The mechanisms may involve inhibiting transcription of inflammatory cytokines and oxidative stress, thus protecting the integrity of the vascular endothelium. Meanwhile VitC reduces shedding of SDC-1 and alters p38/MAPK phosphorylation and microcirculation.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Animals , Ascorbic Acid/pharmacology , Cardiopulmonary Resuscitation/methods , Disease Models, Animal , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Syndecan-1/therapeutic use , p38 Mitogen-Activated Protein Kinases
3.
Crit Care Med ; 50(2): e189-e198, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34637412

ABSTRACT

OBJECTIVES: To investigate the therapeutic potential and underlying mechanisms of exogenous nicotinamide adenine dinucleotide+ on postresuscitation myocardial and neurologic dysfunction in a rat model of cardiac arrest. DESIGN: Thirty-eight rats were randomized into three groups: 1) Sham, 2) Control, and 3) NAD. Except for the sham group, untreated ventricular fibrillation for 6 minutes followed by cardiopulmonary resuscitation was performed in the control and NAD groups. Nicotinamide adenine dinucleotide+ (20 mg/kg) was IV administered at the onset of return of spontaneous circulation. SETTING: University-affiliated research laboratory. SUBJECTS: Sprague-Dawley rats. INTERVENTIONS: Nicotinamide adenine dinucleotide+. MEASUREMENTS AND MAIN RESULTS: Hemodynamic and myocardial function were measured at baseline and within 4 hours following return of spontaneous circulation. Survival analysis and Neurologic Deficit Score were performed up to 72 hours after return of spontaneous circulation. Adenosine triphosphate (adenosine triphosphate) level was measured in both brain and heart tissue. Mitochondrial respiratory chain function, acetylation level, and expression of Sirtuin3 and NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 9 (NDUFA9) in isolated mitochondrial protein from both brain and heart tissue were evaluated at 4 hours following return of spontaneous circulation. The results demonstrated that nicotinamide adenine dinucleotide+ treatment improved mean arterial pressure (at 1 hr following return of spontaneous circulation, 94.69 ± 4.25 mm Hg vs 89.57 ± 7.71 mm Hg; p < 0.05), ejection fraction (at 1 hr following return of spontaneous circulation, 62.67% ± 6.71% vs 52.96% ± 9.37%; p < 0.05), Neurologic Deficit Score (at 24 hr following return of spontaneous circulation, 449.50 ± 82.58 vs 339.50 ± 90.66; p < 0.05), and survival rate compared with that of the control group. The adenosine triphosphate level and complex I respiratory were significantly restored in the NAD group compared with those of the control group. In addition, nicotinamide adenine dinucleotide+ treatment activated the Sirtuin3 pathway, down-regulating acetylated-NDUFA9 in the isolated mitochondria protein. CONCLUSIONS: Exogenous nicotinamide adenine dinucleotide+ treatment attenuated postresuscitation myocardial and neurologic dysfunction. The responsible mechanisms may involve the preservation of mitochondrial complex I respiratory capacity and adenosine triphosphate production, which involves the Sirtuin3-NDUFA9 deacetylation.


Subject(s)
Heart Arrest/complications , Heart Failure/drug therapy , NAD/pharmacology , Nervous System Diseases/drug therapy , Resuscitation/standards , Animals , Disease Models, Animal , Heart Arrest/drug therapy , Heart Failure/prevention & control , NAD/therapeutic use , Nervous System Diseases/prevention & control , Rats , Rats, Sprague-Dawley/injuries , Rats, Sprague-Dawley/metabolism , Resuscitation/methods , Resuscitation/statistics & numerical data
4.
Shock ; 57(3): 344-350, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34618729

ABSTRACT

ABSTRACT: Blocking ferroptosis reduces ischemia-reperfusion injury in some pathological contexts. However, there is no evidence that ferroptosis contributes to post-resuscitation myocardial dysfunction (PRMD). Here, we evaluated the therapeutic performance of ferroptosis inhibitors (UAMC-3203 or/and Deferoxamine) on the PRMD in a rat model of cardiac arrest and surveyed the changes of essential ferroptosis markers in the myocardium. Remarkably, all treatments reduce the severity of cardiac dysfunction and microcirculation hypoperfusion after resuscitation compared with control. Consistently, we observe that the ferroptosis marker Glutathione peroxidase 4, 4-hydroxynonenal and non-heme iron altered (1 ±â€Š0.060 vs. 0.021 ±â€Š0.016, 1 ±â€Š0.145 vs. 3.338 ±â€Š0.221, 52.010 ±â€Š3.587 ug/g vs. 70.500 ±â€Š3.158 ug/g, all P < 0.05) in the myocardium after resuscitation. These changes were significantly suppressed by UAMC-3203 [(0.187 ±â€Š0.043, 2.848 ±â€Š0.169, all P < 0.05), (72.43 ±â€Š4.920 ug/g, P  > 0.05)], or Deferoxamine (0.203 ±â€Š0.025, 2.683 ±â€Š0.273, 55.95 ±â€Š2.497 ug/g, all P < 0.05). Briefly, UAMC-3203 or/and Deferoxamine improve post-resuscitation myocardial dysfunction and provide evidence of ferroptosis involvement, suggesting that ferroptosis inhibitors could potentially provide an innovative therapeutic approach for mitigating the myocardial damage caused by cardiopulmonary resuscitation.


Subject(s)
Cardiopulmonary Resuscitation/adverse effects , Deferoxamine/therapeutic use , Ferroptosis/drug effects , Heart Arrest/therapy , Myocardial Reperfusion Injury/prevention & control , Siderophores/therapeutic use , Animals , Cyclohexylamines/agonists , Disease Models, Animal , Male , Phenylenediamines/agonists , Rats , Rats, Sprague-Dawley
5.
Open Life Sci ; 16(1): 1064-1081, 2021.
Article in English | MEDLINE | ID: mdl-34676301

ABSTRACT

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe syndrome lacking efficient therapy and resulting in high morbidity and mortality. Although resveratrol (RES), a natural phytoalexin, has been reported to protect the ALI by suppressing the inflammatory response, the detailed mechanism of how RES affected the immune system is poorly studied. Pulmonary conventional dendritic cells (cDCs) are critically involved in the pathogenesis of inflammatory lung diseases including ALI. In this study, we aimed to investigate the protective role of RES via pulmonary cDCs in lipopolysaccharide (LPS)-induced ALI mice. Murine ALI model was established by intratracheally challenging with 5 mg/kg LPS. We found that RES pretreatment could mitigate LPS-induced ALI. Additionally, proinflammatory-skewed cytokines decreased whereas anti-inflammatory-related cytokines increased in bronchoalveolar lavage fluid by RES pretreatment. Mechanistically, RES regulated pulmonary cDCs' maturation and function, exhibiting lower level of CD80, CD86, major histocompatibility complex (MHC) II expression, and IL-10 secretion in ALI mice. Furthermore, RES modulated the balance between proinflammation and anti-inflammation of cDCs. Moreover, in vitro RES pretreatment regulated the maturation and function of bone marrow derived dendritic cells (BMDCs). Finally, the adoptive transfer of RES-pretreated BMDCs enhanced recovery of ALI. Thus, these data might further extend our understanding of a protective role of RES in regulating pulmonary cDCs against ALI.

6.
Biomed Pharmacother ; 142: 111935, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34467895

ABSTRACT

The physiology and physiopathology process of mitochondrial function following cardiac arrest remains poorly understood. We aimed to assess mitochondrial respiratory function on the heart and brain homogenates from cardiac arrest rats. The expression level of SIRT1/PGC-1α pathway was measured by immunoblotting. 30 rats were assigned to the CA group and the sham group. Rats of CA were subjected to 6 min of untreated ventricular fibrillation (VF) followed by 8 min of cardiopulmonary resuscitation (CPR). Mitochondrial respiratory function was compromised following CA and I/R injury, as indicated by CIL (451.46 ± 71.48 vs. 909.91 ± 5.51 pmol/min*mg for the heart and 464.14 ± 8.22 vs. 570.53 ± 56.33 pmol/min*mg for the brain), CI (564.04 ± 64.34 vs. 2729.52 ± 347.39 pmol/min*mg for the heart and 726.07 ± 85.78 vs. 1762.82 ± 262.04 pmol/min*mg for the brain), RCR (1.88 ± 0.46 vs. 3.57 ± 0.38 for the heart and 2.05 ± 0.19 vs. 3.49 ± 0.19, for the brain) and OXPHOS coupling efficiency (0.45 ± 0.11 vs. 0.72 ± 0.03 for the heart and 0.52 ± 0.05 vs. 0.71 ± 0.01 for the brain). However, routine respiration was lower in the heart and comparable in the brain after CA. CIV did not change in the heart but was enhanced in the brain. Furthermore, both SIRT1 and PGC-1α were downregulated concurrently in the heart and brain. The mitochondrial respiratory function was compromised following CA and I/R injury, and the major affected respiratory state is complex I-linked respiration. Furthermore, the heart and the brain respond differently to the global I/R injury after CA in mitochondrial respiratory function. Inhibition of the SIRT1/PGC-1α pathway may be a major contributor to the impaired mitochondrial respiratory function.


Subject(s)
Brain/metabolism , Cardiopulmonary Resuscitation , Heart Arrest/metabolism , Heart Arrest/physiopathology , Mitochondria/metabolism , Myocardium/metabolism , Animals , Biological Oxygen Demand Analysis , Disease Models, Animal , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Respiration , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Spirometry , Ventricular Fibrillation/metabolism
7.
J Am Heart Assoc ; 10(9): e019177, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33884887

ABSTRACT

Background To investigate the therapeutic potential of combined therapy with polyethylene glycol-20k (PEG-20k) and MCC950 on post-resuscitation myocardial function in a rat model of cardiac arrest. Methods and Results Thirty rats were randomized into 5 groups: Sham, Control, PEG-20k, MCC950, PEG-20k+ MCC950. Except for sham, animals were subjected to 6 minutes of ventricular fibrillation followed by 8 minutes cardiopulmonary resuscitation. Two milliliters PEG-20k was administered by intravenous injection coincident with the start of cardiopulmonary resuscitation; MCC950 (10 mg/kg), a highly selective NLRP3 inflammasome inhibitor, was delivered immediately after restoration of spontaneous circulation. Myocardial function, sublingual microcirculation, mitochondrial function, plasma cardiac troponin I, and interleukin-1ß, expression of proteins in SIRT1 (sirtuin 1)/PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha) and NLRP3 (the NOD-like receptor family protein 3) inflammasome pathways were evaluated. Following cardiopulmonary resuscitation, myocardial function was compromised with a significantly decreased cardiac output, ejection fraction, and increased myocardial performance index, cardiac troponin I. Sublingual microcirculation was disturbed with impaired perfused vessel density and microvascular flow index. Cardiac arrest reduced mitochondrial routine respiration, Complex I-linked respiration, respiratory control rates and oxidative phosphorylation coupling efficiency. PEG-20k or MCC950 alone restored mitochondrial respiratory function, restituted sublingual microcirculation, and preserved myocardial function, whereas a combination of PEG-20k and MCC950 further improved these aspects. PEG-20k restored the expression of SIRT1 and PGC-1α, and blunted activation of NLRP3 inflammasomes. MCC950 suppressed expression of cleaved-caspase-1/pro-caspase-1, ASC (apoptosis-associated speck-like protein), GSDMD [gasdermin d], and interleukin-1ß. Conclusions Combined therapy with PEG-20k and MCC950 is superior to either therapy alone for preserving post-resuscitated myocardial function, restituting sublingual microcirculation at restoration of spontaneous circulation at 6 hours. The responsible mechanisms involve upregulated expression of SIRT1/PGC1-α in tandem with inhibition of NLRP3 inflammasomes.


Subject(s)
Cardiopulmonary Resuscitation/methods , Furans/pharmacology , Heart Arrest/therapy , Indenes/pharmacology , Inflammasomes/metabolism , Myocardial Contraction/drug effects , Polyethylene Glycols/pharmacology , Stroke Volume/physiology , Sulfonamides/pharmacology , Animals , Disease Models, Animal , Heart Arrest/complications , Heart Arrest/metabolism , Male , Rats , Rats, Sprague-Dawley , Stroke Volume/drug effects , Ventricular Fibrillation/complications
8.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33300050

ABSTRACT

Post­cardiac arrest myocardial dysfunction (PAMD) is a leading cause of death in patients undergoing resuscitation patients following cardiac arrest (CA). Although prostaglandin E1 (PGE1) is a clinical drug used to mitigate ischemia injury, its effect on PAMD remains unknown. In the present study, the protective effects of PGE1 on PAMD were evaluated in a rat model of CA and in a hypoxia­reoxygenation (H/R) in vitro model. Rats were randomly assigned to CA, CA+PGE1 or sham groups. Asphyxia for 8 min followed by cardiopulmonary resuscitation were performed in the CA and CA+PGE1 groups. PGE1 was intravenously administered at the onset of return of spontaneous circulation (ROSC). PGE1 treatment significantly increased the ejection fraction and cardiac output within 4 h following ROSC and improved the survival rate, compared with the CA group. Moreover, PGE1 inactivated GSK3ß, prevented mitochondrial permeability transition pore (mPTP) opening, while reducing cytochrome c and cleaved caspase­3 expression, as well as cardiomyocyte apoptosis in the rat model. To examine the underlying mechanism, H/R H9c2 cells were treated with PGE1 at the start of reoxygenation. The changes in GSK3ß activity, mPTP opening, cytochrome c and cleaved caspase­3 expression, and apoptosis of H9c2 cells were consistent with those noted in vivo. The results indicated that PGE1 attenuated PAMD by inhibiting mitochondria­mediated cardiomyocyte apoptosis.


Subject(s)
Alprostadil/pharmacology , Apoptosis/drug effects , Heart Arrest/metabolism , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Animals , Heart Arrest/drug therapy , Heart Arrest/pathology , Male , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar
9.
Inhal Toxicol ; 32(2): 79-85, 2020 02.
Article in English | MEDLINE | ID: mdl-32188325

ABSTRACT

Objective: Acute carbon monoxide (CO)poisoning can cause delayed neurological sequelae (DNS). Glycogen synthase kinase 3ß (GSK-3ß) /Tau protein pathway is reported to play a key role in neurological abnormalities. In the present study, we aimed to determine the role of GSK-3ß/Tau in DNS following acute CO poisoning.Methods: 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), a specific non-competitive inhibitor of GSK-3ß, was used to inhibit GSK-3ß. Twenty-four male Sprague-Dawley rats were randomly assigned to the three groups: Control group, CO group and CO-TDZD-8 group. Rats breathed 1000 ppm CO for 40 minutes and then 3000 ppm for up to 20 minutes until they lost consciousness. TDZD-8 (1 mg/kg) was administered intravenously three times after the end of CO exposure at 0, 24, 48 hours late. Learning and memory abilities were observed using the Morris Water Maze (MWM). Brain histological changes were evaluated by hematoxylin-eosin staining. Moreover, the expression levels of Tau and GSK-3ß were detected after acute carbon monoxide poisoning.Results: TDZD-8 significantly attenuated the learning and memory dysfunction induced by acute CO poisoning, ameliorated the histology structure of damaged neural cells in cortex and hippocampus CA1 area. TDZD-8 clearly decreased p-Tau expression, reversed the reduction of p-GSK-3ß induced by acute CO poisoning.Conclusions: The therapeutic effect of TDZD-8 in alleviating DNS caused by acute CO poisoning is related to the inactivation of Tau by intensifying the level of GSK-3ß phosphorylation.


Subject(s)
Carbon Monoxide Poisoning/drug therapy , Glycogen Synthase Kinase 3/metabolism , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Thiadiazoles/therapeutic use , tau Proteins/metabolism , Animals , Carbon Monoxide Poisoning/complications , Carbon Monoxide Poisoning/metabolism , Carbon Monoxide Poisoning/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Learning/drug effects , Male , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Thiadiazoles/pharmacology
10.
Basic Clin Pharmacol Toxicol ; 122(5): 470-480, 2018 May.
Article in English | MEDLINE | ID: mdl-29151273

ABSTRACT

Delayed encephalopathy after acute carbon monoxide (CO) poisoning (DEACMP) is the most severe and clinically intractable complication that occurs following acute CO poisoning. Unfortunately, the mechanism of DEACMP is still vague. Growing evidence indicates that delayed cerebral damage after CO poisoning is related to oxidative stress, abnormal neuro-inflammation, apoptosis and immune-mediated injury. Our recent report indicated that methylene blue (MB) may be a promising therapeutic agent in the prevention of neuronal cell death and cognitive deficits after transient global cerebral ischaemia (GCI). In this study, we aimed to investigate the potential of MB therapy to ameliorate the signs and symptoms of DEACMP. Rats were exposed to 1000 ppm CO for 40 min. in the first step; CO was then increased to 3000 ppm, which was maintained for another 20 min. The rats were implanted with 7-day release Alzet osmotic mini-pumps subcutaneously under the back skin, which provided MB at a dose of 0.5 mg/kg/day 1 hr after CO exposure. The results showed that MB significantly suppressed oxidative damage and expression of pro-inflammatory factors, including tumour necrosis factor-α and interleukin (IL)-1ß. MB treatment also suitably modulated mitochondrial fission and fusion, which is helpful in the preservation of mitochondrial function. Furthermore, MB dramatically attenuated apoptosis and neuronal death. Lastly, behavioural studies revealed that MB treatment preserved spatial learning and memory in the Barnes maze test. Our findings indicated that MB may have protective effects against DEACMP.


Subject(s)
Antidotes/pharmacology , Brain Diseases/prevention & control , CA1 Region, Hippocampal/drug effects , Carbon Monoxide Poisoning/drug therapy , Methylene Blue/pharmacology , Neurons/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Behavior, Animal/drug effects , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Diseases/physiopathology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Carbon Monoxide Poisoning/metabolism , Carbon Monoxide Poisoning/physiopathology , Cognition/drug effects , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Male , Maze Learning/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Spatial Learning/drug effects , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
Inflamm Res ; 65(3): 213-24, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26681130

ABSTRACT

INTRODUCTION: Urinary trypsin inhibitor (UTI) decreases inflammatory cytokine levels and mortality in experimental animal models of inflammation. Here, we observed the effect of UTI on lipopolysaccharide (LPS)-induced hyperpermeability in human umbilical vein endothelial cells (HUVECs) and explored the role of vascular endothelial-cadherin (VE-cadherin) in its effect. METHODS: The effect of UTI on endothelial barrier hyperpermeability was detected by an electrical cell-substrate impedance sensing (ECIS) system and a transwell chamber system. The expression of VE-cadherin in HUVECs was examined by real-time PCR and western blot. RESULTS: We demonstrated that the alleviation of LPS-induced barrier dysfunction could be achieved by pretreatment with 3000 U/mL of UTI. VE-cadherin monoclonal antibody (mAb) could inhibit the protective effects. UTI maintained VE-cadherin expression by increasing protein stability at both the transcriptional and post-transcriptional levels. Meanwhile, VE-cadherin expression on the cell surface increased when the cells were pretreated with UTI. Furthermore, pretreatment with UTI decreased the phosphorylation of VE-cadherin at Tyr658 but not Tyr731. CONCLUSIONS: Our data show that prophylactic UTI maintains the endothelial barrier function, increases VE-cadherin expression, and inhibits the phosphorylation of VE-cadherin at Tyr658 under inflammatory conditions. It suggests a scientific and potential clinical therapeutic importance of UTI in treatment of inflammatory disorders.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Glycoproteins/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Antigens, CD/genetics , Cadherins/genetics , Cell Adhesion Molecules/genetics , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Claudins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipopolysaccharides , Macrophages/drug effects , Macrophages/physiology , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Up-Regulation
12.
Inhal Toxicol ; 27(12): 613-20, 2015.
Article in English | MEDLINE | ID: mdl-26444456

ABSTRACT

Acute pulmonary edema is one of the major outcomes of exposure to high levels of hydrogen sulfide (H2S). However, the mechanisms involved in H2S-induced acute pulmonary edema are still poorly understood. Therefore, the present study is designed to evaluate the role of epithelial sodium channel (ENaC) in H2S-induced acute pulmonary edema. The Sprague-Dawley rats were exposed to sublethal concentrations of inhaled H2S, then the pulmonary histological and lung epithelial cell injury were evaluated by hematoxylin-eosin staining and electron microscopy, respectively. In addition to morphological investigation, our results also revealed that H2S exposure significantly decreased the alveolar fluid clearance and increased the lung tissue wet-dry ratio. These changes were demonstrated to be associated with decreased ENaC expression. Furthermore, the extracellular-regulated protein kinases 1/2 pathway was demonstrated to be implicated in H2S-mediated ENaC expression, because PD98059, an ERK1/2 antagonist, significantly mitigated H2S-mediated ENaC down-regulation. Therefore, our results show that ENaC might represent a novel pharmacological target for the treatment of acute pulmonary edema induced by H2S and other hazardous gases.


Subject(s)
Epithelial Sodium Channels/metabolism , Hydrogen Sulfide/toxicity , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/physiology , Humans , Hydrogen Sulfide/administration & dosage , Inhalation Exposure/adverse effects , Pulmonary Edema/pathology , Rats , Rats, Sprague-Dawley
13.
Environ Toxicol Pharmacol ; 38(2): 616-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25195098

ABSTRACT

Acute pulmonary edema (APE) is one of the fatal outcomes after exposure to high levels of hydrogen sulfide (H2S), available evidence suggest that dexamethasone (DXM), a potent anti-inflammatory agent, has been widely used or proposed as a therapeutic approach for H2S-induced APE in clinical practice, however, the underlying mechanism remains poorly understood. Ample evidence suggest that epithelial Na(+) channel, especially for the subunit α-epithelial Na(+) channel (α-ENaC) plays a critical role in alveolar fluid clearance. Therefore, the present study is undertaken to investigate the effects of DXM on α-ENaC following H2S exposure. The Sprague Dawley rats were exposed to H2S to establish APE model, in parallel, A549 cells were treated with NaHS to establish cell model. In vivo study, we found that DXM significantly attenuated H2S-induced lung histopathological changes and alveolar fluid clearance decrement, however, these preventive effects of DXM can be obviously counteracted by the mifepristone (MIF), the glucocorticoid receptor (GR) blocker. Moreover, DXM markedly attenuated H2S-mediated α-ENaC down-regulation, and similarly, the process can be partially retarded by MIF. Furthermore, DXM obviously prevented H2S-mediated ERK1/2 activation both in vitro and in vivo study. These results, taken together, suggested that DXM exerted protective effects on H2S-induced APE, and α-ENaC might be a potential therapeutic target for APE induced by H2S.


Subject(s)
Dexamethasone/administration & dosage , Epithelial Sodium Channels/metabolism , Lung/drug effects , Protective Agents/administration & dosage , Pulmonary Edema/chemically induced , Pulmonary Edema/drug therapy , Animals , Cell Line , Dexamethasone/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Hydrogen Sulfide , Lung/pathology , Male , Mifepristone/administration & dosage , Mifepristone/pharmacology , Protective Agents/pharmacology , Pulmonary Edema/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
14.
PLoS One ; 9(4): e94701, 2014.
Article in English | MEDLINE | ID: mdl-24722316

ABSTRACT

Acute lung injury (ALI) is one of the fatal outcomes after exposure to high levels of hydrogen sulfide (H2S), and the matrix metalloproteinases (MMPs) especially MMP-2 and MMP-9 are believed to be involved in the development of ALI by degrading the extracellular matrix (ECM) of blood-air barrier. However, the roles of MMP-2 and MMP-9 in H2S-induced ALI and the mechanisms of dexamethasone (DXM) in treating ALI in clinical practice are still largely unknown. The present work was aimed to investigate the roles of MMP-2 and MMP-9 in H2S-induced ALI and the protective effects of DXM. In our study, SD rats were exposed to H2S to establish the ALI model and in parallel, A549 cells were incubated with NaHS (a H2S donor) to establish cell model. The lung HE staining, immunohistochemisty, electron microscope assay and wet/dry ratio were used to identify the ALI induced by H2S, then the MMP-2 and MMP-9 expression in both rats and A549 cells were detected. Our results revealed that MMP-2 and MMP-9 were obviously increased in both mRNA and protein level after H2S exposure, and they could be inhibited by MMP inhibitor doxycycline (DOX) in rat model. Moreover, DXM significantly ameliorated the symptoms of H2S-induced ALI including alveolar edema, infiltration of inflammatory cells and the protein leakage in BAFL via up-regulating glucocorticoid receptor(GR) to mediate the suppression of MMP-2 and MMP-9. Furthermore, the protective effects of DXM in vivo and vitro study could be partially blocked by co-treated with GR antagonist mifepristone (MIF). Our results, taken together, demonstrated that MMP-2 and MMP-9 were involved in the development of H2S-induced ALI and DXM exerted protective effects by alleviating the expression of MMP-2 and MMP-9. Therefore, MMP-2 and MMP-9 might represent novel pharmacological targets for the treatment of H2S and other hazard gases induced ALI.


Subject(s)
Acute Lung Injury/drug therapy , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Cell Line , Dexamethasone/pharmacology , Disease Models, Animal , Glucocorticoids/pharmacology , Hydrogen Sulfide , Lung/drug effects , Lung/metabolism , Male , Rats , Rats, Sprague-Dawley
15.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 25(8): 471-4, 2013 Aug.
Article in Chinese | MEDLINE | ID: mdl-24021042

ABSTRACT

OBJECTIVE: To explore the risk factors on prognosis of Acinetobacter baumannii bloodstream infection. METHODS: Clinical data from 78 patients with Acinetobacter baumannii bloodstream infection hospitalized in First Affiliated Hospital of Nanjing Medical University from January 2010 to November 2012 were analyzed retrospectively. According to the 28-day prognosis after admission, the patients were divided into non-survivors (n=40) and survivors (n=38). Data on demographic and clinical characteristics, wards, underlying diseases, treatments, invasive medical procedures, bacterial resistance to antibiotics, and acute physiology and chronic health evaluation II (APACHEII) score in the beginning were collected. The index as an independent risk factor of mortality was demonstrated by multivariate logistic regression analysis. The predictor value was concluded by comparing area under the receiver operating characteristic curve (ROC curve) of each index. RESULTS: Risk factors of mortality of Acinetobacter baumannii bloodstream infection goes as following, including intensive care unit admission[ICU, odds ratio (OR)=12.9,95% confidence interval (95%CI) 2.4-63.5, P=0.001], trachea intubation or tracheostomy (OR=6.2, 95%CI 1.5-30.4, P=0.023), invasive mechanical ventilation (OR=5.1, 95%CI 1.4-22.6, P=0.042), invasive medical procedure besides central venous catheter (including thoracentesis, bone marrow puncture, lumbar puncture, catheterization, bronchoalveolar lavage with bronchofibroscope, arteriovenous fistula plastic operation, OR=8.4, 95%CI 1.7-37.8, P=0.011), APACHEII score ≥19 in the beginning (OR=35.4, 95%CI 3.8-318.6, P=0.001). With respect to APACHE II score≥ 19 as mortality cut-off point, an area under the receiver operating curve of 0.938 was statistically significant (P<0.05), with sensitivity 76.2% and specificity 94.1%. The relationship between prognosis and antibiotic resistance did not have statistically significance. CONCLUSIONS: Invasive medical procedures and treatments were associated with increased mortality of patients with Acinetobacter baumannii bloodstream infection. A strong predictor of adverse outcome in such conditions was APACHEII score ≥19.


Subject(s)
Acinetobacter Infections/diagnosis , Bacteremia/diagnosis , APACHE , Acinetobacter Infections/mortality , Acinetobacter baumannii , Adolescent , Adult , Aged , Aged, 80 and over , Bacteremia/microbiology , Bacteremia/mortality , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Factors , Survival Rate , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...