Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Article in Chinese | MEDLINE | ID: mdl-37667152

ABSTRACT

Objective: To explore the toxic effect of carbon black nanoparticles on human bronchial epithelial cells, and identify the differentially expressed circular RNA based on the full transcriptome high-throughput sequencing, so as to provide evidence for the development of biomarkers exposed to carbon black nanoparticles and their application on epigenetic toxicology. Methods: In June 2020, 16 HBE cells were treated with carbon black nanoparticles at concentrations of 20, 40 and 80 µg/ml, and 16 HBE cells without any intervention were used as the control group. The cytotoxicity of carbon black nanoparticles was detected by CCK8 and LDH experiments. Real-time quantitative fluorescent PCR (qRT-PCR) and ELISA were used to detect the changes of interleukin-6 (IL-6) and interleukin-8 (IL-6, IL-8) mRNA and protein levels of carbon black nanoparticles with concentration gradient after 72 h exposure. Western blot analysis was conducted to detect the expression levels of toll-like receptor 4 (TLR4), phosphorylated nuclear factor-κB (P-NF-κB), apoptosis-related speckled protein (ASC) and Caspase-1 associated with nuclear factor-κB. According to high-throughput sequencing results, differentially expressed Circrnas were screened and identified by qRT-PCR, and those with stable differentially expressed circrnas and the strongest association with the NF-κB pathway were selected for ring performance identification. Results: After being exposed to carbon black nanoparticles for 72 h, the activity of 16HBE cells decreased significantly (P<0.05), and the release of lactate dehydrogenase increased significantly (P<0.05). Compared with control group, mRNA expression levels of IL-6 and IL-8, protein levels of IL-6 and IL-8 were increased, and protein levels of TLR4, p-NF-κB, ASC and Caspase-1 were significantly up-regulated in 16 HBE cells of different concentrations, with statistical significance (P<0.05). Compared with the control group, a total of 492 differentially expressed circular Rnas (|log2 FC|>1) were detected. Among the 5 differentially expressed (P<0.05) circular Rnas, circ_002642 was selected as the object of subsequent research on circular Rnas, affter 72 hours of exposure to 80 µg/ml CBNPs, 16HBE cells showed signlficantly higher expression of circ_002642 (P<0.05) . Conclusion: Carbon black nanoparticles can induce differentially expressed circular RNAs associated with inflammatory response in human bronchial epithelial cells.


Subject(s)
NF-kappa B , RNA, Circular , Humans , Interleukin-8 , Soot/toxicity , Toll-Like Receptor 4 , Interleukin-6 , Epithelial Cells , Caspase 1
2.
Zhonghua Wei Chang Wai Ke Za Zhi ; 25(6): 531-538, 2022 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-35754218

ABSTRACT

Objective: To systematically evaluate the efficacy and safety of total neoadjuvant therapy (TNT) in the comprehensive treatment of locally advanced rectal cancer. Methods: Literatures were screened from PubMed, Embase, Web of Science, Cochrane Library, CBM, Wanfang Data, VIP and CNKI from the inception date to May 2021 to collect the randomized controlled clinical trials (RCTs) of TNT followed by total mesorectal excision (TME) versus neoadjuvant chemotherapy (nCRT) followed by TME in the treatment of locally advanced rectal cancer. The data of overall survival, disease-free survival, R0 radical resection rate, pathological complete response (pCR) rate, T downstaging rate, the incidence of adverse events ≥ grade III, including neutropenia, nausea and vomiting, diarrhea, radiation dermatitis and nervous system toxicity, and the morbidity of complications within postoperative 30 days of the two groups were extracted from the included literatures. Review Manager 5.3 software was utilized for statistical meta-analysis. Results: Nine RCTs were finally enrolled including 2430 patients. Meta-analysis results showed that compared with nCRT group, patients in TNT group had longer overall survival (HR=0.80, 95%CI: 0.65-0.97, P=0.03) and higher pCR rate (RR=1.73, 95%CI: 1.44-2.08, P<0.01) with significant differences. Besides, there were no significant differences between two groups in disease-free survival (HR=0.86, 95%CI:0.71-1.05, P=0.14), R0 radical resection rate (RR=1.02, 95%CI: 0.99-1.06, P=0.17) and T downstaging rate (RR=1.04, 95%CI: 0.89-1.22, P=0.58) between two groups. In terms of treatment safety, the incidence of adverse events ≥ grade III (RR=1.09, 95%CI: 0.70-1.70, P=0.70) and morbidity of complications within postoperative 30 days (RR=1.07, 95%CI: 0.97-1.18, P=0.19) did not significantly differ between two groups. Conclusions: In the treatment of locally advanced rectal cancer, TNT may bring more survival benefits than nCRT and does not increase the incidence of adverse events and postoperative complications. Therefore, TNT could be used as a recommended treatment for patients with locally advanced rectal cancer.


Subject(s)
Neoplasms, Second Primary , Rectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols , Chemoradiotherapy/methods , Disease-Free Survival , Humans , Neoadjuvant Therapy/methods , Neoplasm Staging , Neoplasms, Second Primary/pathology , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Rectum/pathology , Treatment Outcome
3.
Zhonghua Xin Xue Guan Bing Za Zhi ; 49(7): 687-693, 2021 Jul 24.
Article in Chinese | MEDLINE | ID: mdl-34256436

ABSTRACT

Objective: To estimate the diagnostic value of fibronectin type Ⅲ-domain containing protein 5 (FNDC5) in subclinical diabetic cardiomyopathy. Methods: A total of 94 patients with type 2 diabetes (T2DM), who were hospitalized from April 2018 to June 2019 in the Third Affiliated Hospital of Soochow University, were enrolled in this study. Patients were divided into T2DM with cardiac dysfunction (subclinical DCM) group (n=47) and T2DM without cardiac dysfunction (non-DCM) group (n=47) according to echocardiography and gated myocardial perfusion imaging results. Basic clinical data and serum FNDC5 level were compared between the two groups. Logistic regression analysis was used to establish predicting models and the diagnostic efficiency of established models was compared by ROC curve analysis. Results: Compared to non-DCM group, patients in subclinical DCM group were older, with longer duration of diabetes, and had higher levels of glycosylated hemoglobin A1c (HbA1c), total cholesterol (TC), triglyceride (TG) and low-density lipoprotein cholesterol (LDL-C) (all P<0.05). Serum FNDC5 level was significantly lower in subclinical DCM group than in non-DCM group (P<0.001). FNDC5 level was positively correlated with ventricular septal e'(r=0.451,P=0.005), mitral valve e'(r=0.291,P<0.001), the ratio of peak early diastolic trans-mitral flow velocity (E) to peak late diastolic trans-mitral flow velocity (A)(r=0.490,P=0.002), while negatively correlated with A(r=-0.399,P<0.001), the average ratio of E/e'(r=-0.490,P<0.001), tricuspid regurgitation velocity(r=-0.567,P<0.001), left atrial volume index(r=-0.491,P<0.001). Univariate ROC analysis showed that the diagnostic efficacy of FNDC5(AUC=0.940,95%CI 0.897-0.982)was superior to age(AUC=0.639,95%CI 0.523-0.752), diabetic duration(AUC=0.663,95%CI 0.555-0.772), HbA1c(AUC=0.740,95%CI 0.638-0.839), TG(AUC=0.661,95%CI 0.547-0.776), TC(AUC=0.675,95%CI 0.563-0.788)and LDL-C(AUC=0.644,95%CI 0.532-0.756). Model 1 was established with subclinical DCM as dependent variable, age, diabetic duration, TG, TC, LDL-C and HbA1c as independent variables. Model 2 was established by adding FNDC5 as independent variable on the basis of model 1. Diagnostic efficacy for subclinical DCM was compared between the two models by ROC analysis. The diagnostic efficiency was better with model 2 (AUC=0.980) than with model 1 (AUC=0.879, P<0.001). When sensitivity was set at 0.617, the specificity of model 2 was higher than that of model 1(0.979 vs. 0.936). When sensitivity was set at 0.532, the sensitivity of model 2 was higher than that of model 1 (1.000 vs. 0.915). Conclusions: Our findings suggest that serum FNDC5 could be used as a novel biomarker for the diagnosis of subclinical DCM.

5.
Genet Mol Res ; 15(4)2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27966751

ABSTRACT

Cinnamyl alcohol dehydrogenase (CAD) catalyzes the final step in lignin biosynthesis. The genus Eucalyptus belongs to the family Myrtaceae, which is the main cultivated species in China. Eucalyptus urophylla GLU4 (GLU4) is widely grown in Guangxi. It is preferred for pulping because of its excellent cellulose content and fiber length. Based on GLU4 and CAD gene expression, a Eucalyptus variety low in lignin content should be obtained using transgenic technology, which could reduce the cost of pulp and improve the pulping rate, and have favorable prospects for application. However, the role and function of CAD in GLU4 is still unclear. In the present study, EuCAD was cloned from GLU4 and identified using bioinformatic tools. Subsequently, in order to evaluate its impact on lignin synthesis, a full-length EuCAD RNAi vector was constructed, and transgenic tobacco was obtained via Agrobacterium-mediated transformation. A significant decrease in CAD expression and lignin content in transgenic tobacco demonstrated a key role for EuCAD in lignin biosynthesis and established a regulatory role for RNAi. In our study, the direct molecular basis of EuCAD expression was determined, and the potential regulatory effects of this RNAi vector on lignin biosynthesis in E. urophylla GLU4 were demonstrated. Our results provide a theoretical basis for the study of lignin biosynthesis in Eucalyptus.


Subject(s)
Alcohol Oxidoreductases/genetics , Cloning, Molecular/methods , Eucalyptus/enzymology , Nicotiana/genetics , Alcohol Oxidoreductases/metabolism , China , Eucalyptus/genetics , Gene Expression Regulation, Plant , Lignin/biosynthesis , Phylogeny , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified/growth & development , Nicotiana/growth & development
6.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 34(12): 931-934, 2016 Dec 20.
Article in Chinese | MEDLINE | ID: mdl-28241684

ABSTRACT

Objective: To investigate the features of peripheral nerve injuries in workers exposed to vibration. Methods: A total of 197 male workers [median age: 34 years (21-50 years) ; median working years of vibration exposure: 7.3 years (1-20 years) ] engaged in grinding in an enterprise were enrolled. Their clinical data and electromyography results were analyzed to investigate the features of peripheral nerve impairment. Results: Of all workers, 96 (48.73%) had abnormal electromyography results. Of all workers, 88 (44.7%) had simple mild median nerve injury in the wrist, who accounted for 91.7% (88/96) of all workers with abnormal electromy-ography results. Six workers had ulnar nerve injury, superficial radial nerve injury, or/and superficial peroneal nerve injury and accounted for 6.3% of all workers with abnormal electromyography results. Of all workers, 88 had a reduced amplitude of median nerve sensory transduction, and 28 had slowed median nerve sensory transduction. A total of 46 workers were diagnosed with occupational hand-arm vibration disease and hospitalized for treatment. They were followed up for more than 4 months after leaving their jobs, and most of them showed improvements in neural electromyography results and returned to a normal state. Conclusion: Workers exposed to vibration have a high incidence rate of nerve injury in the hand, mainly sensory function impairment at the distal end of the median nerve, and all injuries are mild peripheral nerve injuries. After leaving the vibration job and being treated, most workers can achieve improvements and return to a normal state.


Subject(s)
Hand-Arm Vibration Syndrome , Peripheral Nerve Injuries/etiology , Vibration/adverse effects , Adult , Carpal Tunnel Syndrome , Electromyography , Hand , Humans , Male , Median Nerve , Middle Aged , Neural Conduction , Occupational Diseases , Occupations , Peripheral Nerves , Physical Therapy Modalities , Wrist , Young Adult
7.
Cell Death Dis ; 6: e1624, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25633293

ABSTRACT

DRAM1 (DNA damage-regulated autophagy modulator 1) is a TP53 target gene that modulates autophagy and apoptosis. We previously found that DRAM1 increased autophagy flux by promoting lysosomal acidification and protease activation. However, the molecular mechanisms by which DRAM1 regulates apoptosis are not clearly defined. Here we report a novel pathway by which DRAM1 regulates apoptosis involving BAX and lysosomes. A549 or HeLa cells were treated with the mitochondrial complex II inhibitor, 3-nitropropionic acid (3NP), or an anticancer drug, doxorubicin. Changes in the protein and mRNA levels of BAX and DRAM1 and the role of DRAM1 in BAX induction were determined. The interaction between DRAM1 and BAX and its effect on BAX degradation, BAX lysosomal localization, the release of cathepsin B and cytochrome c by BAX and the role of BAX in 3NP- or doxorubicin-induced cell death were studied. The results showed that BAX, a proapoptotic protein, was induced by DRAM1 in a transcription-independent manner. BAX was degraded by autophagy under basal conditions; however, its degradation was inhibited when DRAM1 expression was induced. There was a protein interaction between DRAM1 and BAX and this interaction prolonged the half-life of BAX. Furthermore, upregulated DRAM1 recruited BAX to lysosomes, leading to the release of lysosomal cathepsin B and cleavage of BID (BH3-interacting domain death agonist). BAX mediated the release of mitochondrial cytochrome c, activation of caspase-3 and cell death partially through the lysosome-cathepsin B-tBid pathway. These results indicate that DRAM1 regulates apoptosis by inhibiting BAX degradation. In addition to mitochondria, lysosomes may also be involved in BAX-initiated apoptosis.


Subject(s)
Apoptosis , Lysosomes/metabolism , Membrane Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Autophagy , BH3 Interacting Domain Death Agonist Protein/metabolism , Cathepsin B/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Humans , Models, Biological , Protein Binding , Protein Transport , Proteolysis , Transcription, Genetic
8.
Neuroscience ; 284: 900-911, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25446361

ABSTRACT

BACKGROUND: Rotenone is an environmental neurotoxin that induces accumulation of α-synuclein and degeneration of dopaminergic neurons in substantia nigra pars compacta (SNpc), but the molecular mechanisms are not fully understood. We investigated whether rotenone induced impairment of autophagic flux and lysosomal functions. METHODS: Autophagy flux, accumulation of α-synuclein, lysosomal membrane integrity and neurodegeneration were assessed in the rotenone-treated rat model and PC12 cells, and the effects of the autophagy inducer trehalose on rotenone's cytotoxicity were also studied. RESULTS: Rotenone administration significantly reduced motor activity and caused a loss of tyrosine hydroxylase in SNpc of Lewis rats. The degeneration of nigral dopaminergic neurons was accompanied by the deposition of α-synuclein aggregates, autophagosomes and redistribution of cathepsin D from lysosomes to the cytosol. In cultured PC12 cells, rotenone also induced increases in protein levels of α-synuclein, microtubule-associated protein 1 light chain 3-II, Beclin 1, and p62. Rotenone increased lysosomal membrane permeability as evidenced by leakage of N-acetyl-beta-d-glucosaminidase and cathepsin D, the effects were blocked by reactive oxygen species scavenger tiron. Autophagy inducer trehalose enhanced the nuclear translocation of transcription factor EB, accelerated the clearance of autophagosomes and α-synuclein and attenuated rotenone-induced cell death of PC12 cells. Meanwhile, administration of trehalose to rats in drinking water (2%) decreased rotenone-induced dopaminergic neurons loss in SNpc. CONCLUSIONS: These studies indicate that the lysosomal dysfunction contributes to rotenone's neurotoxicity and restoration of lysosomal function could be a new therapeutic strategy for Parkinson's disease.


Subject(s)
Autophagy/drug effects , Lysosomes/drug effects , Parkinsonian Disorders/physiopathology , Rotenone/toxicity , Animals , Antiparkinson Agents/pharmacology , Autophagy/physiology , Lysosomes/metabolism , Male , PC12 Cells , Parkinsonian Disorders/drug therapy , Pars Compacta/drug effects , Pars Compacta/physiopathology , Random Allocation , Rats , Rats, Inbred Lew , Trehalose/pharmacology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
9.
Neuroscience ; 207: 52-64, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22330834

ABSTRACT

The present study sought to investigate if p53 mediates autophagy activation and mitochondria dysfunction in primary striatal neurons in kainic acid (KA)-induced excitotoxicity. The excitotoxic model of primary striatal neurons was established with KA. The levels of p53, microtubule-associated protein 1 light chain 3 (LC3), Beclin1, and p62 were examined by Western blot and immunostaining. Autophagy activation was also determined with electron microscope. To evaluate the contribution of p53 to autophagy activation and mitochondria dysfunction in KA-induced excitotoxicity, the protein levels of LC3, Beclin1, and p62, the mitochondrial transmembrane potential and the mitochondrial Reactive oxygen species (ROS) after pretreatment with the p53 inhibitor pifithrin-alpha (PFT-α) and the autophagy inhibitor 3-methyladenine (3-MA) were analyzed. Excitotoxic neuronal injury was induced after KA treatment as demonstrated by increases in lactate dehydrogenase (LDH) leakage and was significantly inhibited by PFT-α. Western blot and immunostaining showed that the induction of p53 protein occurred in the cytosol and the nucleus. Increases in autophagic proteins LC3 and Beclin1 were observed, whereas the protein levels of p62 decreased after KA treatment. Electron microscope analysis showed increased autophagosomes in the cytoplasm. The changes in LC3, Beclin1, and p62 levels were blocked by PFT-α, PFT-µ, 3-MA, and E64d but not Z-DEVD-FMK. JC-1 staining showed the depolarization of mitochondrial membrane potential after excitotoxic insult. Mito-tracker and RedoxSensor Red CC-1 staining showed an increased production of mitochondrial ROS after excitotoxic insult. These effects were significantly suppressed after pretreatment with PFT-α and 3-MA. This study suggests that p53 mediates KA-induced autophagy activation and mitochondrial dysfunction in striatal neurons.


Subject(s)
Autophagy/physiology , Corpus Striatum/pathology , Mitochondrial Diseases/pathology , Neurons/pathology , Neurotoxins/toxicity , Tumor Suppressor Protein p53/physiology , Animals , Autophagy/drug effects , Corpus Striatum/metabolism , Kainic Acid/toxicity , Mitochondrial Diseases/metabolism , Neurons/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/antagonists & inhibitors
10.
Oncogene ; 31(8): 1055-64, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-21743489

ABSTRACT

Nucleus accumbens-1 (NAC1), a nuclear factor belonging to the BTB/POZ gene family, is known to have important roles in proliferation and growth of tumor cells and in chemotherapy resistance. Yet, the mechanisms underlying how NAC1 contributes to drug resistance remain largely unclear. We report here that autophagy was involved in NAC1-mediated resistance to cisplatin, a commonly used chemotherapeutic drug in the treatment of ovarian cancer. We found that treatment with cisplatin caused an activation of autophagy in ovarian cancer cell lines, A2780, OVCAR3 and SKOV3. We further demonstrated that knockdown of NAC1 by RNA interference or inactivation of NAC1 by inducing the expression of a NAC1 deletion mutant that contains only the BTB/POZ domain significantly inhibited the cisplatin-induced autophagy, resulting in increased cisplatin cytotoxicity. Moreover, inhibition of autophagy and sensitization to cisplatin by NAC1 knockdown or inactivation were accompanied by induction of apoptosis. To confirm that the sensitizing effect of NAC1 inhibition on the cytotoxicity of cisplatin was attributed to suppression of autophagy, we assessed the effects of the autophagy inhibitors 3-methyladenosine and chloroquine, and small interfering RNAs (siRNAs) targeting beclin 1 or Atg5 on the cytotoxicity of cisplatin. Treatment with 3-methyladenosine, chloroquine or beclin 1 and Atg5-targeted siRNA also enhanced the sensitivity of SKOV3, A2780 and OVCAR3 cells to cisplatin, indicating that suppression of autophagy indeed renders tumor cells more sensitive to cisplatin. Regulation of autophagy by NAC1 was mediated by the high-mobility group box 1 (HMGB1), as the functional status of NAC1 was associated with the expression, translocation and release of HMGB1. The results of our study not only revealed a new mechanism determining cisplatin sensitivity but also identified NAC1 as a novel regulator of autophagy. Thus, the NAC1-mediated autophagy may be exploited as a new target for enhancing the efficacy of cisplatin against ovarian cancer and other types of malignancies.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Autophagy/drug effects , Cisplatin/pharmacology , HMGB1 Protein/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chloroquine/pharmacology , Drug Resistance, Neoplasm , Female , Gene Expression , Gene Knockdown Techniques , HMGB1 Protein/genetics , Humans , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Ovarian Neoplasms , Protein Transport , RNA Interference , Repressor Proteins/genetics , Sequestosome-1 Protein
11.
Neuroscience ; 184: 54-63, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21463664

ABSTRACT

Previous data demonstrate that traumatic brain injury (TBI) activates autophagy, and increases microtubule-associated protein 1 light chain 3 (LC3) immunostaining mainly in neurons. However, the role of autophagy in traumatic brain damage remains elusive. The aim of the present study was to investigate the autophagic mechanisms participating in traumatic brain injury. The autophagy inhibitors 3-methyladenine (3-MA) and bafliomycin A1 (BFA) were administered with a single i.c.v. injection before TBI. We first examined the protein levels of Beclin-1 and LC3 II, which have been found to promote autophagy previously. Immunoblotting analysis showed that 3-MA pretreatment reduced post-TBI Beclin-1 and LC3-II levels, and maintained p62/SQSTM1 (p62) levels. In addition, double immunolabeling showed that the increased punctate LC3-II dots colocalizing with Propidium Iodide (PI)-stained nuclei at 24 h after injury, were partially inhibited by 3-MA pretreatment. Furthermore, inhibition of autophagy could reduce TBI-induced cell injury assessed with i.p. injection of PI and lesion volume, and attenuate behavioral outcome evaluated by motor test and Morris water maze. The neuroprotective effects were associated with an inhibition on TBI-induced up-regulation of LC3, Beclin-1, cathepsin B, caspase-3 and the Beclin-1/Bcl-2 ratio. Taken together, these data imply that the autophagy pathway is involved in the pathophysiologic responses after TBI, and inhibition of this pathway may help attenuate traumatic damage and functional outcome deficits.


Subject(s)
Autophagy/physiology , Brain Injuries/physiopathology , Cell Death/physiology , Maze Learning/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Injuries/metabolism , Brain Injuries/pathology , Male , Mice , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/physiology
12.
Neuroscience ; 176: 381-95, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21185913

ABSTRACT

Previous studies have demonstrated that pioglitazone (Piog), a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, inhibits ischemia-induced brain injury. Piog has also been shown to exert anti-inflammatory effects by attenuation of nuclear factor-κB (NF-κB) activation after myocardial ischemia/reperfusion injury. Because NF-κB is known to play a major role in the pathophysiology of brain ischemia, the present study was undertaken to elucidate whether pioglitazone attenuates ischemic neuronal damage through PPARγ-mediated suppression of NF-κB apoptotic signaling pathway. Permanent middle cerebral artery occlusion (pMCAO) model was induced by using an intraluminal filament technique in rats. Piog was administrated i.p. twice (24 h before and at the time of ischemia insult) or once (10 min after ischemia). The neuroprotection of Piog was analyzed by assessing neurological deficits, infarction volume and morphological changes. The inhibition of NF-κB signaling pathway by Piog was evaluated by detecting the nuclear translocation of NF-κB p65 with immunohistochemistry and its target gene p53 by real-time PCR, and the expression of phospholated NF-κB p65 (p- NF-κB p65) in primary cultured neurons and the protein levels of IκBα and p-ERK in the ischemic cortex or striatum with Western blotting analysis. The contribution of a PPARγ mechanism to Piog's inhibitory effects on NF-κB and neuroprotection was evaluated by pretreatment with the PPARγ irreversible antagonist GW9662. In vitro ischemia in cultured primary neurons was induced by the oxygen-glucose deprivation (OGD) and the protective effect of Piog on cultured neurons was measured by lactate dehydrogenase (LDH) assay. Piog (0.5, 1, 2 mg/kg) reduced infarction volume, and improved morphological changes and motor deficits. Piog markedly up-regulated the protein levels of IκBα or p-ERK 6 h or 12 h after ischemia. Piog reduced the nuclear translocation of NF-κB p65 in the ischemic cortical cells and repressed the expression of p53 12 h after ischemia. Pre-treatment with GW9662 blocked Piog-elicited reduction in infarction volume, the increase in protein levels of IκBα and p-ERK, the reduction in the nuclear translocation of NF-κB subunit p65 and the repression of p53 mRNA expression. In addition, Piog attenuated the OGD-induced neuronal damage and inhibited the OGD-induced increases in p- NF-κB p65 in neurons. The present findings suggest that Piog's neuroprotection appears to be associated with PPARγ-mediated suppression of NF-κB signaling pathway.


Subject(s)
Infarction, Middle Cerebral Artery/metabolism , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , PPAR gamma/metabolism , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pioglitazone , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
13.
J Phys Condens Matter ; 21(13): 134019, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-21817494

ABSTRACT

Nucleation, growth and thermal stability of Pt particles supported on well ordered Fe(3)O(4)(111) thin films grown on Pt(111) were studied by scanning tunnelling microscopy (STM) and temperature programmed desorption (TPD) of CO. STM studies showed that Pt grows through the formation of single-layer islands that coalesce at high coverage. Vacuum annealing at 600 K caused Pt sintering and the formation of extended two-dimensional (2D) islands one and two layers in thickness at sub-monolayer coverage. Well faceted, three-dimensional (3D) Pt nanoparticles formed by annealing to temperatures above 800 K were encapsulated by an FeO(111) monolayer. These results were rationalized in terms of the high adhesion energy for Pt on iron oxide surfaces. CO TPD studies showed that 2D structures, formed at 600 K, exhibit much lower CO adsorption capacity as compared to the Pt(111) single crystal surface. This effect has been tentatively assigned to lattice expansion in the Pt 2D islands leading to weakening of the Pt-CO bond due to reduction of the [Formula: see text] back-donation.

14.
Neuroscience ; 153(2): 414-27, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18400410

ABSTRACT

To define mechanisms underlying neurovascular injury following brain embolism-induced neurodegeneration, we investigated temporal and spatial pathological changes in brain microvessels up to 12 weeks after microsphere embolism (ME) induction in aged male rats. Mild ME upregulated endothelial nitric oxide synthase (eNOS) and protein tyrosine nitration in brain microvessels. Strong beta-amyloid immunoreactivity coincident with increased eNOS immunoreactivity was observed in microvessels. Immunoblotting of purified brain microvessels revealed that beta-amyloid accumulation significantly increased 1 week after ME induction and remained elevated for 12 weeks. Importantly, beta-amyloid accumulation in brain parenchyma was also observed in areas surrounding injured microvessels at 12 weeks. Levels of Alzheimer's-related hyperphosphorylated tau proteins also concomitantly increased in neurons surrounding regions of beta-amyloid accumulation 12 weeks after ME induction, as did glycogen synthase kinase (GSK3beta) (Tyr-216) phosphorylation. Taken together, ME-induced aberrant eNOS expression and subsequent protein tyrosine nitration in microvessels preceded beta-amyloid accumulation both in microvessels and brain parenchyma, leading to hyperphosphorylation of neuronal tau proteins through GSK3beta activation.


Subject(s)
Aging/physiology , Amyloid beta-Peptides/metabolism , Brain Chemistry/physiology , Embolism/metabolism , Microspheres , tau Proteins/metabolism , Animals , Blotting, Western , Capillaries/metabolism , Cerebrovascular Circulation/physiology , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Glycogen Synthase Kinase 3/metabolism , Immunohistochemistry , Male , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Peroxynitrous Acid/metabolism , Phosphorylation , Poly Adenosine Diphosphate Ribose/metabolism , Rats , Rats, Wistar
15.
Neuroscience ; 151(3): 671-9, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18164824

ABSTRACT

The bilateral olfactory bulbectomy (OBX) mouse exhibits neurodegeneration of cholinergic neurons in the medial septum with concomitant cognitive deficits. Consistent with our previous observations, choline acetyltransferase (ChAT) protein levels in the medial septum decreased by 43.5% 2 weeks after OBX without changes in glutamic acid decarboxylase-65 (GAD65) levels. Interestingly, levels of the vesicular acetylcholine transporter (VAChT), which is localized at cholinergic neuron terminals, decreased both in hippocampal CA1 and CA3 regions following OBX. Confocal microscopy showed that VAChT expression was more severely reduced in CA3 14 days after OBX compared with CA1. Intriguingly, chronic treatment with a vanadium (IV) compound, VO(OPT) [bis(1-N-oxide-pyridine-2-thiolato)oxovanadium(IV)] (0.5-1 mg as vanadium (V)/kg/day, i.p.), significantly rescued cholinergic neurons in the medial septum in a dose-dependent manner. VO(OPT) treatment also prevented decreased VAChT immunoreactivity both in CA1 and CA3 regions in the hippocampus. Consistent with these findings, an impaired hippocampal long-term potentiation (LTP) and memory deficits seen in OBX mice were significantly prevented by VO(OPT) treatment. Taken together, OBX induces neurodegeneration of septo-hippocampal cholinergic neurons and impairment of memory-related behaviors. The neuroprotective effect of VO(OPT) could lead to novel therapeutic strategies to ameliorate cognitive deficits associated with cholinergic neuron degeneration in Alzheimer's disease and other neurodegenerative disorders.


Subject(s)
Acetylcholine/metabolism , Hippocampus/pathology , Nerve Degeneration , Neurons/drug effects , Olfactory Bulb/injuries , Organometallic Compounds/therapeutic use , Animals , Behavior, Animal/drug effects , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Male , Maze Learning/drug effects , Mice , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Neurons/metabolism , Olfactory Bulb/metabolism , Patch-Clamp Techniques , Vesicular Acetylcholine Transport Proteins/metabolism
16.
J Forensic Sci ; 50(1): 173-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15831015

ABSTRACT

A few papers in the literature reported incident deaths by acute ketamine poisoning. In this paper, we report an unusual homicide caused by chronic ketamine poisoning. The victim was a 34-year old married woman with no previous medical history (except as reported herein) who died in her own home. The court investigation revealed that she was chronically poisoned by her husband over a period of about one year in an act of homicide. Determination of ketamine concentrations in autopsy specimens was carried out with gas-chromatography/mass spectrometry (GC-MS). The results showed that ketamine concentration was 21 microg/mL in gastric contents, 3.8 microg/mL in blood and 1.2 microg/mL in urine. The most striking forensic findings were cardiac muscle fibrosis and hyaline degeneration of small arteries in victim's heart, the pathological features of ketamine poisoning previous reported only in animal studies.


Subject(s)
Anesthetics, Dissociative/poisoning , Homicide , Ketamine/poisoning , Myocardium/pathology , Adult , Autopsy , Chronic Disease , Female , Fibrosis , Humans
17.
Proc Natl Acad Sci U S A ; 98(22): 12784-9, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11675509

ABSTRACT

The Huntington's disease (HD) mutation is a polyglutamine expansion in the N-terminal region of huntingtin (N-htt). How neurons die in HD is unclear. Mutant N-htt aggregates in neurons in the HD brain; expression of mutant N-htt in vitro causes cell death. Other in vitro studies show that proteolysis by caspase 3 could be important in regulating mutant N-htt function, but there has been no direct evidence for caspase 3-cleaved N-htt fragments in brain. Here, we show that N-htt fragments consistent with the size produced by caspase 3 cleavage in vitro are resident in the cortex, striatum, and cerebellum of normal and adult onset HD brain and are similar in size to the fragments seen after exogenous expression of human huntingtin in mouse clonal striatal neurons. HD brain extracts treated with active caspase 3 had increased levels of N-htt fragments. Compared with the full-length huntingtin, the caspase 3-cleaved N-htt fragments, especially the mutant fragment, preferentially segregated with the membrane fraction. Partial proteolysis of the human caspase 3-cleaved N-htt fragment by calpain occurred in vitro and resulted in smaller N-terminal products; products of similar size appeared when mouse brain protein extracts were treated with calpain. Results support the idea that sequential proteolysis by caspase 3 and calpain may regulate huntingtin function at membranes and produce N-terminal mutant fragments that aggregate and cause cellular dysfunction in HD.


Subject(s)
Brain/metabolism , Calpain/physiology , Caspases/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Adult , Animals , Caspase 3 , Cell Membrane/metabolism , Humans , Huntingtin Protein , Huntington Disease/therapy , Mice , Mutation
18.
Neuroscience ; 106(3): 603-12, 2001.
Article in English | MEDLINE | ID: mdl-11591460

ABSTRACT

Huntington's disease is a progressive, inherited neurodegenerative disorder characterized by the loss of subsets of neurons primarily in the striatum. In this study, we assessed the neuroprotective effect of lithium against striatal lesion formation in a rat model of Huntington's disease in which quinolinic acid was unilaterally infused into the striatum. For this purpose, we used a dopamine receptor autoradiography and glutamic acid decarboxylase mRNA in situ hybridization analysis, methods previously shown to be adequate for quantitative analysis of the excitotoxin-induced striatal lesion size. Here we demonstrated that subcutaneous injections of LiCl for 16 days prior to quinolinic acid infusion considerably reduced the size of quinolinic acid-induced striatal lesion. Furthermore, these lithium pre-treatments also decreased the number of striatal neurons labeled with the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. Immunohistochemistry and western blotting demonstrated that lithium-elicited neuroprotection was associated with an increase in Bcl-2 protein levels. Our results raise the possibility that lithium may be considered as a neuroprotective agent in treatment of neurodegenerative diseases such as Huntington's disease.


Subject(s)
Cell Death/drug effects , Huntington Disease/drug therapy , Lithium/pharmacology , Neostriatum/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Animals , Antimanic Agents/pharmacology , Benzazepines/pharmacokinetics , Cell Death/physiology , Cyclin D1/drug effects , Cyclin D1/metabolism , Disease Models, Animal , Dopamine Antagonists/pharmacokinetics , Glutamate Decarboxylase/genetics , Huntington Disease/chemically induced , Huntington Disease/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Isoenzymes/genetics , Lithium Chloride/pharmacology , Male , Neostriatum/pathology , Neostriatum/physiopathology , Neurons/metabolism , Neurons/pathology , Quinolinic Acid/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/metabolism
19.
Biol Trace Elem Res ; 81(2): 177-83, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11554398

ABSTRACT

A multitracer solution obtained from the nuclear reaction of selenium with 25-MeV/nucleon 40Ar ions was orally administered to normal and tumor-bearing Balb/c male mice. After 96 h, the mice were sacrificed and the elemental distribution was determined in various tissues, organs, and blood. The uptake of Na, Rb, Ga, Sc, V, Cr, Mn, Co, Fe, Zn, Y, Zr, Tc, Ru, Ag, and In in normal and, except for zinc, in tumor-bearing mice was simultaneously detected. Most elements were distributed in about the same manner in the skin and liver of animals in both groups. The distribution of Rb, Ga, V, Cr, Tc, and In showed little or no significant differences between the two study groups. The distribution of Na, Mn, Fe, Ag, Sc, and Co showed significant differences between normal and tumor-bearing mice. In the blood, spleen, and kidney of the normal mice, there was good absorption of Na, Mn, Fe, Ag, Co, and Zn. In the heart, these elements were well absorbed, except for Na and Mn.


Subject(s)
Trace Elements/metabolism , Animals , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Radioactive Tracers , Spectrophotometry , Time Factors , Tissue Distribution
20.
J Pharmacol Exp Ther ; 297(1): 78-87, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259530

ABSTRACT

Prostaglandin A(1) (PGA1) reportedly inhibits NF-kappaB activation and induces expression of heat shock proteins. Since both these effects could be neuroprotective, the therapeutic potential of PGA1 in neurodegenerative disorders, where excitotoxicity may contribute to pathogenesis, was evaluated in rat striatal neurons exposed to the N-methyl-D-aspartate (NMDA) receptor agonist quinolinic acid (QA). Intrastriatal administration of PGA1 (5-80 nmol) attenuated QA (60 nmol)-induced internucleosomal DNA fragmentation. The inhibitory effects of a single dose of PGA1 (80 nmol) on QA (60 nmol)-induced DNA fragmentation were observed 12 to 48 h after treatment. PGA1 (80 nmol) also attenuated QA-induced DNA fragmentation when administered up to 4 h after QA exposure. PGA1 significantly decreased the loss of D1 dopamine receptors and GAD(67) mRNA in QA-injected striatum as measured by quantitative receptor autoradiography and in situ hybridization histochemistry, suggesting that it reduced the neuronal loss induced by QA. Protection of striatal neurons against QA-induced death by PGA1 was further indicated by Nissl staining 10 days after QA administration. PGA1 (5-80 nmol) significantly inhibited QA-induced NF-kappaB activation by blocking inhibitory kappaB-alpha degradation but had no effect on activator protein-1 binding activity. PGA1 (80 nmol) treatment substantially increased 70- and 72-kDa heat shock protein levels in striatum. These results indicate that PGA1 blunts NMDA receptor-mediated neuronal apoptosis by a mechanism possibly involving the up-regulation of neuroprotective heat shock proteins and inhibition of NF-kappaB activation. In view of its potent neuroprotective activity, PGA1 could prove useful in the treatment of certain neurodegenerative disorders related to excitotoxicity.


Subject(s)
Corpus Striatum/drug effects , Prostaglandins A/pharmacology , Quinolinic Acid/toxicity , Animals , Apoptosis/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA Fragmentation/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nucleosomes/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...