Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Fish Shellfish Immunol ; 149: 109564, 2024 Jun.
Article En | MEDLINE | ID: mdl-38631439

Grass carp reovirus (GCRV) infections and hemorrhagic disease (GCHD) outbreaks are typically seasonally periodic and temperature-dependent, yet the molecular mechanism remains unclear. Herein, we depicted that temperature-dependent IL-6/STAT3 axis was exploited by GCRV to facilitate viral replication via suppressing type Ⅰ IFN signaling. Combined multi-omics analysis and qPCR identified IL-6, STAT3, and IRF3 as potential effector molecules mediating GCRV infection. Deploying GCRV challenge at 18 °C and 28 °C as models of resistant and permissive infections and switched to the corresponding temperatures as temperature stress models, we illustrated that IL-6 and STAT3 expression, genome level of GCRV, and phosphorylation of STAT3 were temperature dependent and regulated by temperature stress. Further research revealed that activating IL-6/STAT3 axis enhanced GCRV replication and suppressed the expression of IFNs, whereas blocking the axis impaired viral replication. Mechanistically, grass carp STAT3 inhibited IRF3 nuclear translocation via interacting with it, thus down-regulating IFNs expression, restraining transcriptional activation of the IFN promoter, and facilitating GCRV replication. Overall, our work sheds light on an immune evasion mechanism whereby GCRV facilitates viral replication by hijacking IL-6/STAT3 axis to down-regulate IFNs expression, thus providing a valuable reference for targeted prevention and therapy of GCRV.


Carps , Fish Diseases , Interferon Type I , Interleukin-6 , Reoviridae Infections , Reoviridae , STAT3 Transcription Factor , Signal Transduction , Virus Replication , Animals , Fish Diseases/immunology , Fish Diseases/virology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Reoviridae/physiology , Carps/immunology , Carps/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Interferon Type I/immunology , Interferon Type I/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Immunity, Innate/genetics
2.
Chin J Traumatol ; 2024 Mar 16.
Article En | MEDLINE | ID: mdl-38548574

PURPOSE: Although traditional craniotomy (TC) surgery has failed to show benefits for the functional outcome of intracerebral hemorrhage (ICH). However, a minimally invasive hematoma removal plan to avoid white matter fiber damage may be a safer and more feasible surgical approach, which may improve the prognosis of ICH. We conducted a historical cohort study on the use of multimodal image fusion-assisted neuroendoscopic surgery (MINS) for the treatment of ICH, and compared its safety and effectiveness with traditional methods. METHODS: This is a historical cohort study involving 241 patients with cerebral hemorrhage. Divided into MINS group and TC group based on surgical methods. Multimodal images (CT skull, CT angiography, and white matter fiber of MRI diffusion-tensor imaging) were fused into 3 dimensional images for preoperative planning and intraoperative guidance of endoscopic hematoma removal in the MINS group. Clinical features, operative efficiency, perioperative complications, and prognoses between 2 groups were compared. Normally distributed data were analyzed using t-test of 2 independent samples, Non-normally distributed data were compared using the Kruskal-Wallis test. Meanwhile categorical data were analyzed via the Chi-square test or Fisher's exact test. All statistical tests were two-sided, and p < 0.05 was considered statistically significant. RESULTS: A total of 42 patients with ICH were enrolled, who underwent TC surgery or MINS. Patients who underwent MINS had shorter operative time (p < 0.001), less blood loss (p < 0.001), better hematoma evacuation (p = 0.003), and a shorter stay in the intensive care unit (p = 0.002) than patients who underwent TC. Based on clinical characteristics and analysis of perioperative complications, there is no significant difference between the 2 surgical methods. Modified Rankin scale scores at 180 days were better in the MINS than in the TC group (p = 0.014). CONCLUSIONS: Compared with TC for the treatment of ICH, MINS is safer and more efficient in cleaning ICH, which improved the prognosis of the patients. In the future, a larger sample size clinical trial will be needed to evaluate its efficacy.

3.
Sheng Li Xue Bao ; 75(1): 99-107, 2023 Feb 25.
Article Zh | MEDLINE | ID: mdl-36859839

Silent information regulator 1 (SIRT1) is one of the seven mammalian proteins of the sirtuin family of NAD+-dependent deacetylases. SIRT1 plays a pivotal role in neuroprotection and ongoing research has uncovered a mechanism by which SIRT1 may exert a neuroprotective effect on Alzheimer's disease (AD). Growing evidence demonstrates that SIRT1 regulates many pathological processes including amyloid-ß precursor protein (APP) processing, neuroinflammation, neurodegeneration, and mitochondrial dysfunction. SIRT1 has recently received enormous attention, and pharmacological or transgenic approaches to activate the sirtuin pathway have shown promising results in the experimental models of AD. In the present review, we delineate the role of SIRT1 in AD from a disease-centered perspective and provides an up-to-date overview of the SIRT1 modulators and their potential as effective therapeutics in AD.


Alzheimer Disease , Sirtuins , Animals , Amyloid beta-Protein Precursor , Animals, Genetically Modified , Sirtuin 1 , Humans
4.
EBioMedicine ; 72: 103591, 2021 Oct.
Article En | MEDLINE | ID: mdl-34563924

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease with high mortality, however with no effective therapy available. METHODS: The effect of favipiravir (FPV) in treating SFTS was evaluated by an integrated analysis on data collected from a single-arm study (n=428), a surveillance study (n=2350) and published data from a randomized controlled trial study (n=145). A 1:1 propensity score matching was performed to include 780 patients: 390 received FPV and 390 received supportive therapy only. Case fatality rates (CFRs), clinical progress, and adverse effects were compared. FINDINGS: FPV treatment had significantly reduced CFR from 20.0% to 9.0% (odds ratio 0.38, 95% confidence interval 0.23-0.65), however showing heterogeneity when patients were grouped by age, onset-to-admission interval, initial viral load and therapy duration. The effect of FPV was significant only among patients aged ≤70 years, with onset-to-admission interval ≤5 days, therapy duration ≥5 days or baseline viral load ≤1 × 106 copies/mL. Age-stratified analysis revealed no benefit in the aging group >70 years, regardless of their sex, onset-to-admission interval, therapy duration or baseline viral load. However, for both ≤60 and 60-70 years groups, therapy duration and baseline viral load differentially affected FPV therapy efficiency. Hyperuricemia and thrombocytopenia, as the major adverse response of FPV usage, were observed in >70 years patients. INTERPRETATION: FPV was safe in treating SFTS patients but showed no benefit for those aged >70 years. Instant FPV therapy could highly benefit SFTS patients aged 60-70 years. FUNDING: China Natural Science Foundation (No. 81825019, 82073617 and 81722041) and China Mega-project for Infectious Diseases (2018ZX10713002 and 2015ZX09102022).


Amides/adverse effects , Amides/therapeutic use , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Pyrazines/adverse effects , Pyrazines/therapeutic use , Severe Fever with Thrombocytopenia Syndrome/drug therapy , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome , Viral Load/drug effects
5.
Signal Transduct Target Ther ; 6(1): 145, 2021 04 16.
Article En | MEDLINE | ID: mdl-33859168

Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) is an emerging tick-borne virus with high fatality and an expanding endemic. Currently, effective anti-SFTSV intervention remains unavailable. Favipiravir (T-705) was recently reported to show in vitro and in animal model antiviral efficacy against SFTSV. Here, we conducted a single-blind, randomized controlled trial to assess the efficacy and safety of T-705 in treating SFTS (Chinese Clinical Trial Registry website, number ChiCTR1900023350). From May to August 2018, laboratory-confirmed SFTS patients were recruited from a designated hospital and randomly assigned to receive oral T-705 in combination with supportive care or supportive care only. Fatal outcome occurred in 9.5% (7/74) of T-705 treated patients and 18.3% (13/71) of controls (odds ratio, 0.466, 95% CI, 0.174-1.247). Cox regression showed a significant reduction in case fatality rate (CFR) with an adjusted hazard ratio of 0.366 (95% CI, 0.142-0.944). Among the low-viral load subgroup (RT-PCR cycle threshold ≥26), T-705 treatment significantly reduced CFR from 11.5 to 1.6% (P = 0.029), while no between-arm difference was observed in the high-viral load subgroup (RT-PCR cycle threshold <26). The T-705-treated group showed shorter viral clearance, lower incidence of hemorrhagic signs, and faster recovery of laboratory abnormities compared with the controls. The in vitro and animal experiments demonstrated that the antiviral efficacies of T-705 were proportionally induced by SFTSV mutation rates, particularly from two transition mutation types. The mutation analyses on T-705-treated serum samples disclosed a partially consistent mutagenesis pattern as those of the in vitro or animal experiments in reducing the SFTSV viral loads, further supporting the anti-SFTSV effect of T-705, especially for the low-viral loads.


Amides/administration & dosage , Antiviral Agents/administration & dosage , Phlebovirus/metabolism , Pyrazines/administration & dosage , Severe Fever with Thrombocytopenia Syndrome/drug therapy , Administration, Oral , Animals , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Prospective Studies , Severe Fever with Thrombocytopenia Syndrome/blood , Severe Fever with Thrombocytopenia Syndrome/genetics , Severe Fever with Thrombocytopenia Syndrome/mortality , Single-Blind Method
6.
Clin Infect Dis ; 63(10): 1288-1294, 2016 Nov 15.
Article En | MEDLINE | ID: mdl-27553371

BACKGROUND: During 2014-2015, an outbreak of Ebola virus disease (EVD) swept across parts of West Africa. No approved antiviral drugs are available for Ebola treatment currently. METHODS: A retrospective clinical case series was performed for EVD patients in Sierra Leone-China Friendship Hospital. Patients with confirmed EVD were sequentially enrolled and treated with either World Health Organization (WHO)-recommended supportive therapy (control group) from 10 to 30 October, or treated with WHO-recommended therapy plus favipiravir (T-705) from 1 to 10 November 2014. Survival and virological characteristics were observed for 85 patients in the control group and 39 in the T-705 treatment group. RESULTS: The overall survival rate in the T-705 treatment group was higher than that of the control group (56.4% [22/39] vs 35.3% [30/85]; P = .027). Among the 35 patients who finished all designed endpoint observations, the survival rate in the T-705 treatment group (64.8% [11/17]) was higher than that of the control group (27.8% [5/18]). Furthermore, the average survival time of the treatment group (46.9 ± 5.6 days) was longer than that of the control group (28.9 ± 4.7 days). Most symptoms of patients in the treatment group improved significantly. Additionally, 52.9% of patients who received T-705 had a >100-fold viral load reduction, compared with only 16.7% of patients in the control group. CONCLUSIONS: Treatment of EVD with T-705 was associated with prolonged survival and markedly reduced viral load, which makes a compelling case for further randomized controlled trials of T-705 for treating EVD.


Amides/therapeutic use , Antiviral Agents/therapeutic use , Ebolavirus , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/mortality , Pyrazines/therapeutic use , Adolescent , Adult , Female , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Kaplan-Meier Estimate , Male , Retrospective Studies , Sierra Leone/epidemiology , Viral Load , Young Adult
7.
Ying Yong Sheng Tai Xue Bao ; 23(8): 2107-13, 2012 Aug.
Article Zh | MEDLINE | ID: mdl-23189686

From December 2008 to October 2009, a seasonal investigation was conducted on the phytoplankton' s community structure and its relationships with environmental factors in Datong Lake. With the comparison of the historical data in 1960, the potential effects of intensive aquaculture on the aquatic environment were analyzed, aimed to provide theoretical support for the sustainable fishery of freshwater lakes. A total of 98 phytoplankton species belonging to 7 phyla and 54 genera were collected, among which, Peridinium bipes, Chroomonas acuta, Chlorella vulgaris, Crytomonas ovate, Cyclotella meneghiniana, Crytomonas erosa, Anabaena circinalis, Microcystis aeruginosa, and Anabaena azotica were the dominant species, and had obvious seasonal variations. The mean annual cell density of the phytoplankton was 1.84 x 10(6) cells x L(-1), being the highest in summer (16.4 x 10(6) cells x L(-1)) and ranged from 1.71 x 10(6) to 1.98 x 10(6) cells x L(-1) in the other three seasons. The values of the abundance index, Shannon index, and Pielou index of the phytoplankton community were 2.01-4.55, 1.26-2.69, and 0.69-1.27, respectively. Canonical correlation analysis (CCA) showed that water depth, water temperature, transparency, and water total phosphorus content, oxidation-reduction potential, and electrical conductivity were the main environmental factors affecting the phytoplankton community structure in the Lake.


Aquaculture , Ecosystem , Lakes , Phytoplankton/growth & development , China , Lakes/analysis , Phosphorus/analysis , Phytoplankton/classification , Population Dynamics , Seasons
8.
Neurotox Res ; 21(4): 368-78, 2012 May.
Article En | MEDLINE | ID: mdl-22147269

Our previous study has shown that acteoside, an antioxidative phenylethanoid glycoside, protect against beta-amyloid (Aß)-induced cytotoxicity in vitro. However, the precise protective mechanisms remains unclear. Heme oxygenase-1 (HO-1) is a crucial factor in the response to oxidative injury, protecting neurons against Aß-induced injury. In the present study we examined to determine whether acteoside upregulates HO-1 expression, and thereby protects PC12 cells against Aß-induced cell death. It was revealed that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We showed that acteoside increased HO-1 expression in vitro and in vivo. Acteoside treatment resulted in nuclear translocation of the transcription factor NF-E2-related factor 2 (Nrf2). Acteoside activated both ERK and PI3 K/Akt, and treatments with the specific ERK inhibitor PD98059, the PI3 K inhibitor LY294002, and the specific Nrf2 siRNA suppressed the acteoside-induced HO-1 expression. The HO-1 inhibitor ZnPP, PD98059, and LY294002 markedly abolished the neuroprotective effect of acteoside against Aß-induced neurotoxicity. Taken together, these results demonstrate that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We also showed that acteoside increased HO-1 expression through activation of ERK and PI3 K/Akt signal pathways in vitro. Upregulation of HO-1 by acteoside may involve in the neuroprotection against Aß-induced neurotoxicity.


Amyloid beta-Peptides/antagonists & inhibitors , Glucosides/pharmacology , Heme Oxygenase-1/biosynthesis , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Phenols/pharmacology , Signal Transduction/drug effects , Amyloid beta-Peptides/toxicity , Animals , Cell Death/drug effects , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucosides/antagonists & inhibitors , Heme Oxygenase-1/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Nerve Degeneration/chemically induced , PC12 Cells , Phenols/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Up-Regulation/drug effects
9.
Zhonghua Wai Ke Za Zhi ; 50(12): 1126-30, 2012 Dec.
Article Zh | MEDLINE | ID: mdl-23336493

OBJECTIVE: Systematic reviews of diagnostic value of the nuclear matrix protein 22 (NMP22) and urine cytology for bladder cancer. METHODS: Development of inclusion criteria, exclusion criteria and search strategy to retrieve relevant literature. Screening the literature according to inclusion criteria and exclusion criteria. Quality evaluation of the screening and data extraction, using MetaDiSc 1.4 software for Meta analysis. RESULTS: In total, 266 relevant studies were searched, excluded 256 studies, and then 10 studies were included, with 4895 patients involved. The pooled sensitivity and specificity of NMP22 to detect bladder cancer were 0.76 (95%CI: 0.74 - 0.77), 0.80 (95%CI: 0.79 - 0.82), respectively. The pooled sensitivity and specificity of urine cytology were 0.36 (95%CI: 0.34 - 0.38), 0.94 (95%CI: 0.93 - 0.95), respectively. The area under curve (AUC) for NMP22 and urine cytology were 0.8533 and 0.8628, and Q(*) index were 0.7863 and 0.7934, respectively. CONCLUSIONS: For the diagnosis of bladder cancer, the sensitivity of NMP22 was higher than urine cytology, but the specificity was lower than urine cytology. Overall diagnostic performance of NMP22 was medium, it was no significant difference with urine cytology. It can't replace urine cytology now.


Nuclear Proteins/analysis , Urinalysis , Urinary Bladder Neoplasms/diagnosis , Cytological Techniques , Humans , Sensitivity and Specificity
10.
Neurosci Bull ; 27(3): 163-72, 2011 Jun.
Article En | MEDLINE | ID: mdl-21614099

OBJECTIVE: Munc18-1 has an important role in neurotransmitter release, and controls every step in the exocytotic pathway in the central nervous system. In the present study, whether epileptic seizure causes a change of Munc18 localization in neuronal nuclei was analyzed. METHODS: Epilepsy models were established by injection of kainic acid (KA) solution into hippocampus of Sprague-Dawley (SD) rats or intraperitoneal injection of KA in Kunming mice. The hippocampal neurons were prepared from embryonic day 18 SD rats, and cultured in neurobasal medium, followed by treatment with glutamate for 3 h. Neuronal and glial nuclei of hippocampus were separated by sucrose density gradient centrifugation. The nucleus-enriched fractions were stained with 0.1% Cresyl Violet for morphological assay. Immunochemistry and immunoelectron microscopy with anti-Munc18-1 antibody were used to determine the nuclear localization of Munc18-1. Immunoblotting was used to detect the protein level of Munc18-1. RESULTS: The localization of Munc18-1 in nucleus of rat hippocampal neuron was confirmed by immunochemistry, immunoelectron microscopy, and immunoblotting detection of neuronal nucleus fraction. In animals receiving intrahippocampal or intraperitoneal injection of KA, immunostaining revealed that the expression of Munc18-1 decreased in pyramidal cell layer of CA regions, as well as in hilus and granular cell layer of dentate gyrus in hippocampus. Moreover, immunoblotting analysis showed that the expression level of Munc18-1 in nucleus fraction of hippocampus significantly decreased in KA-treated animals. The relationship between the change of Munc18-1 expression in neuronal nuclei and neuronal over-activation was also tested in primary cultured neurons. After treatment with 50 µmol/L glutamate acid for 3 h, Munc18-1 level was decreased in nucleus fraction and increased in cytoplasmic fraction of primary cultured neurons. CONCLUSION: These results suggest that excitatory stimulation can induce the distribution change of Munc18-1 in neuron, which may subsequently modulate neuronal functions in brain.


Cell Nucleus/metabolism , Epilepsy/metabolism , Munc18 Proteins/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Epilepsy/chemically induced , Excitatory Amino Acid Agonists , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Kainic Acid , Male , Mice , Mice, Inbred Strains , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley
11.
Brain Res ; 1360: 159-67, 2010 Nov 11.
Article En | MEDLINE | ID: mdl-20828541

Astaxanthin (ATX), the most abundant flavonoids in propolis, has been proven to exert neuroprotective property against glutamate-induced neurotoxicity and ischemia-reperfusion-induced apoptosis. Previous study have revealed that ATX can rescue PC12 cells from Aß(25-35)-induced apoptotic death. However, the mechanisms by which ATX mediates its therapeutic effects in vitro are unclear. In the present study, we explored the underlying mechanisms involved in the protective effects of ATX on the Aß(25-35)-induced cytotoxicity in SH-SY5Y cells. Pre-treatment with ATX for 4h significantly reduced the Aß(25-35)-induced viability loss, apoptotic rate and attenuated Aß-mediated ROS production. In addition, ATX inhibited Aß(25-35)-induced lowered membrane potential, decreased Bcl-2/Bax ratio. We also demonstrated that ATX could prevent the activation of p38MAPK kinase pathways induced by Aß. Moreover, we for the first time have revealed the ATX increased antioxidant enzyme heme oxygenase-1 (HO-1) expression in concentration-dependent and time-dependent manners, which were correlated with its protective effect against Aß(25-35)-induced injury. Because the inhibitor of HO-1 activity, ZnPP reversed the protective effect of ATX against Aß(25-35)-induced cell death. We also demonstrated that the specific ERK inhibitor, PD98059, concentration-dependently blocked on ATX-induced HO-1 expression, and meanwhile PD98059 reversed the protective effect of ATX against Aß25-35-induced cell death. Taken together, these findings suggest that astaxanthin can induce HO-1 expression through activation of ERK signal pathways, thereby protecting the SH-SY5Y cells from Aß(25-35)-induced oxidative cell death.


Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Heme Oxygenase-1/biosynthesis , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Neuroprotective Agents , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Apoptosis Regulatory Proteins/biosynthesis , Blotting, Western , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Flavonoids/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Humans , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Oxidative Stress/drug effects , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Xanthophylls/antagonists & inhibitors , Xanthophylls/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Neurosci Bull ; 26(4): 273-81, 2010 Aug.
Article En | MEDLINE | ID: mdl-20651809

OBJECTIVE: Munc18 is considered as an intracellular protein that plays an important role in exocytosis of neurotransmitters. Previous studies have demonstrated the presence of autoantibodies against Munc18 in a subgroup of Rasmussen's encephalitis patients. However, the machinery of Munc18 autoimmunity is still elusive. The present study was aimed to investigate Munc18 release from primary cultured neurons, Munc18 distribution on the outer plasma membrane of neurons, and the neurotoxicity of Munc18 antibody. METHODS: The cerebral cortical neurons from embryonic day 17 Sprague-Dawley rats were prepared and cultured in neurobasal medium. The proteins in culture medium were precipitated with 10 % trichloroacetic acid, and analyzed by immunoblotting. The proteins on neuronal surface were biotinylated with EZ-Link-sulfo-NHS-LC-Biotin, and collected with avidin-conjugated agarose beads followed by immunoblotting analysis. For cell surface immunofluorescent staining, the living neurons were labeled with anti-Munc18 antibody at 4 degrees C. Neuronal injury was assessed by lactate dehydrogenase(LDH) release. RESULTS: Munc18 was detected in culture medium by immunoblotting analysis. After treatment with 50 micromol/L glutamate for 1 h, Munc18 content in medium was increased. Meanwhile, beta-actin and syntaxin1 were not detected in culture medium, and LDH release was not significantly increased. Moreover, glutamate enhanced Munc18 distribution on outer plasma membrane. Living neuron staining also demonstrated the localization of Munc18 on neuronal surface after glutamate treatment, especially at contacting regions between neurons. Glutamate-induced increase of surface Munc18 distribution was suppressed by NMDA receptor antagonist MK801, but not by AMPA receptor antagonist NBQX. Moreover, compared with c-Fos antibody, Munc18 antibody could induce neuronal injury, when culture medium contained the components of serum. CONCLUSION: A portion of Munc18 can be released from neurons or distributed on neuronal surface, which can be enhanced by glutamate treatment via activation of NMDA receptors. Besides, Munc18 antibody-induced neuronal injury depends on the serum components.


Cerebral Cortex/cytology , Glutamic Acid/pharmacology , Munc18 Proteins/metabolism , Neurons/drug effects , Animals , Biotinylation/methods , Cell Death/drug effects , Cells, Cultured , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , L-Lactate Dehydrogenase/metabolism , Protein Transport/drug effects , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
13.
Brain Res ; 1347: 132-41, 2010 Aug 06.
Article En | MEDLINE | ID: mdl-20513368

Microtubule-associated proteins (MAPs) play a critical role in maintaining normal cytoskeletal architecture and functions. In the present study, we aim to explore the effects of the emotional stressor, chronic restraint stress, on the expression levels and localization of tau and MAP2. We found that after chronic restraint stress, soluble hyperphosphorylated tau was greatly increased, whereas MAP2 was decreased. Moreover, immunohistochemistry analysis demonstrated that phosphorylated tau and MAP2 displayed the similar subcellular distribution pattern after chronic restraint stress. Robust hyperphosphorylated tau immunolabeling was observed both in cortex and hippocampus of stressed animals and mainly located to perikaryal/dendritic elements. After stress, the MAP2 was mainly distributed in the perikaryal compartments, discontinuous dendrites and neuropil. Moreover, the distribution pattern of MAP2 in hippocampus significantly changed. Immunofluorescence double labeling indicated that hyperphosphorylated tau increased in the regions where there displayed an decrease of MAP2. These results suggest that the involvement of repeated restraint stress may not only induce phosphorylation state of tau and distribution of phosphorylated tau, but also alter the content and neuronal distribution of MAP2. Tau and MAP2 are most important MAPs for neuronal cells, the subcellular distribution change of them might be link to functional change of neurons after emotional stress.


Cerebral Cortex/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Microtubule-Associated Proteins/metabolism , Stress, Psychological/pathology , tau Proteins/metabolism , Animals , Disease Models, Animal , Male , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Restraint, Physical/methods , Stress, Psychological/etiology
14.
Biochem Biophys Res Commun ; 390(4): 1294-8, 2009 Dec 25.
Article En | MEDLINE | ID: mdl-19878659

Huntington's disease (HD) and other polyglutamine (polyQ) neurodegenerative diseases are characterized by neuronal accumulation of the disease protein, suggesting that the cellular ability to handle abnormal proteins is compromised. As a multi-subunit protein localized in the mitochondria of eukaryotic cells, the F(0)F(1)-ATP synthase alpha belongs to the family of stress proteins HSP60. Currently, mounting evidences indicate F(0)F(1)-ATP synthase alpha may play a role in neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Recently, ATP synthase alpha was reported to have protective and therapeutic roles in primary cardiacmyocytes of iron-overloaded rats by lowering ROS production. However, little is understood about the role of ATP synthase alpha in cell death and neurodegeneration. Here, we demonstrate that overexpression of ATP synthase alpha suppresses huntingtin (htt) polyQ aggregation and toxicity in transfected SH-SY5Y cell lines. Overexpression of ATP synthase alpha is able to protect cell death caused by polyglutamine-expanded htt. Transient overexpression of ATP synthase alpha suppresses the aggregate formation by estimation of polyQ aggregation, Western blot analysis, and filter trap assay (FTA) in transfected SH-SY5Y cells. These results indicated that ATP synthase alpha has a strong inhibitory effect on polyglutamine aggregate formation and toxicity in vitro, and suggest a novel neuroprotective role of ATP synthase alpha.


Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proton-Translocating ATPases/biosynthesis , Animals , Apoptosis , Cell Line, Tumor , Humans , Huntingtin Protein , Mice , Peptides/pharmacology , Rats
15.
Neurosci Lett ; 465(3): 276-81, 2009 Nov 20.
Article En | MEDLINE | ID: mdl-19766167

The biological function of full-length amyloid-beta protein precursor (APP), the precursor of Abeta, is not fully understood. Mounting studies reported that antibody binding to cell surface APP causes neuronal injury. However, the mechanism of cell surface APP mediating neuronal injury remains to be determined. Colocalization of APP with integrin on cell surface leads us to suppose that focal adhesion (FA) related mechanism is involved in surface APP-mediated neuronal injury. In the present study, results demonstrated that primary cultured neurons treated with antibody against APP-N-terminal not only caused neuronal injury and aberrant morphologic changes of neurite, but also induced reaction of FA proteins appearing an acute increase then decrease pattern. Moreover, the elevation of tyrosine phosphorylation of FA proteins including paxillin and focal adhesion kinase (FAK), and down-regulated expression of protein tyrosine phosphatase (PTP1B) induced by APP antibody were prevented by inhibitor of Src protein kinases 4-amino-5-(4-chlorophenyl)-7(t-butyl) pyrazol (3,4-D) pyramide (PP2) and G protein inhibitor pertussis toxin (PTX), implying that Src family kinase and G protein play roles in APP-induced FA signals. In addition, pretreatment with PTX and PP2 was able to suppress APP-antibody induced neuronal injury. Taken together, the results suggest a novel mechanism for APP mediating neuronal injury through deregulating FA signals.


Amyloid beta-Protein Precursor/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Focal Adhesion Protein-Tyrosine Kinases/immunology , Neurons/immunology , Neurons/pathology , Signal Transduction/immunology , Animals , Apoptosis/drug effects , Cells, Cultured , Neurons/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/immunology , Rats , Signal Transduction/drug effects
16.
Cell Biol Int ; 32(10): 1230-7, 2008 Oct.
Article En | MEDLINE | ID: mdl-18675923

beta-Amyloid protein (Abeta), a major protein component of brain senile plaques in Alzheimer's disease, is known to be directly responsible for the production of reactive oxygen species (ROS) and induction of apoptosis. In this study, the protective effect of puerarin, an isoflavone purified from the radix of the Chinese herb Pueraria lobata, on Abeta-induced rat pheochromocytoma (PC12) cultures was investigated. Although exposure of PC12 cells to 50 microM Abeta25-35 caused significant viability loss and apoptotic rate increase, pretreatment of the cells with puerarin for 24h reduced the viability loss and apoptotic rate. Puerarin (1 microM) significantly inhibited Abeta25-35-induced apoptosis of PC12 cells. Preincubation of the cell with puerarin also restored the ROS and mitochondrial membrane potential levels that had been altered as a result of Abeta25-35 treatment. Puerarin was also found to increase the Bcl-2/Bax ratio and reduce caspase-3 activation. These results suggest that puerarin could attenuate Abeta25-35-induced PC12 cell injure and apoptosis and could also promote the survival of PC12 cells. Therefore, puerarin may act as an intracellular ROS scavenger, and its antioxidant properties may protect against Abeta25-35-induced cell injury.


Amyloid beta-Peptides/pharmacology , Isoflavones/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Caspase 3/metabolism , Humans , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/drug effects , Neurons/physiology , PC12 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Vasodilator Agents/pharmacology , bcl-2-Associated X Protein/metabolism
17.
Zhong Yao Cai ; 31(4): 543-6, 2008 Apr.
Article Zh | MEDLINE | ID: mdl-18661826

OBJECTIVE: To establish a nerve cell injury model by incubating PC12 cell line in the presence of Abeta25-35 to study the effect of puerarin on apoptosis of nerve cells. METHODS: PC12 cells were incubated with Abeta25-35 and puerarin. Cell viability was detected by MTT. The cellular morphology was observed with electron microscopy. FITC-labeled Annexin V and propidium iodide were adopted to evaluate the rate of cell apoptosis in different groups by means of flow cytometry. RESULTS: Following incubation Abeta25-35, the cells were induced to undergo apoptosis. The viability of PC12 cells decreased in a time-dependent manner. Morphological evidences for apoptosis nuclear condensation were observed in PC12 cells. Cells incubated in the presence of Abeta25-35 showed increasing apoptotic rates, but cells treated with puerarin and Abeta25-35 revealed decreasing apoptotic rates, it demonstrated that puerarin had a significant protective action against Abeta25-35 evoked apoptosis. CONCLUSION: Puerarin can resist the adverse effects of Abeta25-35 on increasing apoptotic rates, and it has the protective function towards nerve cells.


Alzheimer Disease/pathology , Apoptosis/drug effects , Isoflavones/pharmacology , Neuroprotective Agents/pharmacology , Amyloid beta-Peptides , Animals , Cell Survival/drug effects , Fabaceae/chemistry , Flow Cytometry , PC12 Cells , Peptide Fragments , Rats , Vasodilator Agents/pharmacology
...