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










Publication year range
1.
J Cardiovasc Dev Dis ; 10(5)2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37233158

ABSTRACT

Type 2 diabetes (T2D) confers a high risk of heart failure frequently with evidence of cardiovascular structural and functional abnormalities before symptom onset. The effects of remission of T2D on cardiovascular structure and function are unknown. The impact of the remission of T2D, beyond weight loss and glycaemia, on cardiovascular structure and function and exercise capacity is described. Adults with T2D without cardiovascular disease underwent multimodality cardiovascular imaging, cardiopulmonary exercise testing and cardiometabolic profiling. T2D remission cases (Glycated hemoglobin (HbA1c) < 6.5% without glucose-lowering therapy, ≥3 months) were propensity score matched 1:4 based on age, sex, ethnicity and time of exposure to those with active T2D (n = 100) with the nearest-neighbour method and 1:1 with non-T2D controls (n = 25). T2D remission was associated with a lower leptin-adiponectin ratio, hepatic steatosis and triglycerides, a trend towards greater exercise capacity and significantly lower minute ventilation/carbon dioxide production (VE/VCO2 slope) vs. active T2D (27.74 ± 3.95 vs. 30.52 ± 5.46, p < 0.0025). Evidence of concentric remodeling remained in T2D remission vs. controls (left ventricular mass/volume ratio 0.88 ± 0.10 vs. 0.80 ± 0.10, p < 0.025). T2D remission is associated with an improved metabolic risk profile and ventilatory response to exercise without concomitant improvements in cardiovascular structure or function. There is a requirement for continued attention to risk factor control for this important patient population.

3.
Heliyon ; 5(9): e02390, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31517118

ABSTRACT

Glioblastoma (GBM) are lethal primary brain tumours whose pathogenesis is aided, at least partly, via a pro-tumorigenic microenvironment. This study investigated whether microglia, a cell component of the GBM microenvironment, mediates pro-tumorigenic properties via the action of cyclophilin A (CypA), a potent secretable chemokine and cytoprotectant that signals via the cell surface receptor, CD147. To this end, intracellular and secreted CypA expression was assessed in human primary microglia and BV2 microglial cells treated with the endotoxin, lipopolysaccharide (LPS) and the oxidative stress inducer, LY83583. We report that human primary microglia and BV2 microglia both express CypA and CD147, and that BV2 microglial cells secrete CypA in response to pro-inflammatory and oxidative stimuli. We also demonstrate for the first time that recombinant CypA (rCypA; 1nM-1000nM) dose-dependently increased wound healing and reduced basal cell death in BV2 microglial cells. To determine the cell-signalling pathways involved, we probed microglial cell lysates for changes in ERK1/2 and AKT phosphorylation, IκB degradation, and IL-6 secretion using Western blot and ELISA analysis. In summary, BV2 microglial cells secrete CypA in response to inflammatory and oxidative stress, and that rCypA increases cell viability and chemotaxis. Our findings suggest that rCypA is a pro-survival chemokine for microglia that may influence the GBM tumour microenvironment.

4.
Medicine (Baltimore) ; 95(40): e5085, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27749584

ABSTRACT

Prostate cancer is one of the most prevalent cancers in men. It is critical to identify and characterize oncogenes that drive the pathogenesis of human prostate cancer. The current study builds upon previous research showing that a disintegrin and metallproteinase (ADAM)28 is involved in the pathogenesis of numerous cancers. Our novel study used overexpression, pharmacological, and molecular approaches to investigate the biological function of ADAM28 in human prostate cancer cells, with a focus on cell proliferation and migration. The results of this study provide important insights into the role of metalloproteinases in human prostate cancer.The expression of ADAM28 protein levels was assessed within human prostate tumors and normal adjacent tissue by immunohistochemistry. Immunocytochemistry and western blotting were used to assess ADAM28 protein expression in human prostate cancer cell lines. Functional assays were conducted to assess proliferation and migration in human prostate cancer cells in which ADAM28 protein expression or activity had been altered by overexpression, pharmacological inhibition, or by siRNA gene knockdown.The membrane bound ADAM28 was increased in human tumor biopsies and prostate cancer cell lines. Pharmacological inhibition of ADAM28 activity and/or knockdown of ADAM28 significantly reduced proliferation and migration of human prostate cancer cells, while overexpression of ADAM28 significantly increased proliferation and migration.ADAM28 is overexpressed in primary human prostate tumor biopsies, and it promotes human prostate cancer cell proliferation and migration. This study supports the notion that inhibition of ADAM28 may be a potential novel therapeutic strategy for human prostate cancer.


Subject(s)
ADAM Proteins/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Prostatic Neoplasms/genetics , ADAM Proteins/biosynthesis , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
5.
Cell Mol Neurobiol ; 34(3): 379-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24395206

ABSTRACT

Using proteomics, we identified nucleoside diphosphate kinase A (NDPKA; also known as NME/NM23 nucleoside diphosphate kinase 1: NME1) to be up-regulated in primary cortical neuronal cultures by erythropoietin (EPO) preconditioning. To investigate a neuroprotective role of NDPKA in neurons, we used a RNAi construct to knock-down and an adenoviral vector to overexpress the protein in cortical neuronal cultures prior to exposure to three ischemia-related injury models; excitotoxicity (L-glutamic acid), oxidative stress (hydrogen peroxide), and in vitro ischemia (oxygen-glucose deprivation). NDPKA down-regulation had no effect on neuronal viability following injury. By contrast, NDPKA up-regulation increased neuronal survival in all three-injury models. Similarly, treatment with NDPKA recombinant protein increased neuronal survival, but only against in vitro ischemia and excitotoxicity. These findings indicate that the NDPKA protein may confer a neuroprotective advantage following injury. Furthermore, as exogenous NDPKA protein was neuroprotective, it suggests that a cell surface receptor may be activated by NDPKA leading to a protective cell-signaling response. Taken together both NDPKAs intracellular and extracellular neuroprotective actions suggest that the protein is a legitimate therapeutic target for the design of drugs to limit neuronal death following stroke and other forms of brain injury.


Subject(s)
Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Nucleoside-Diphosphate Kinase/biosynthesis , Up-Regulation/physiology , Animals , Brain Ischemia/prevention & control , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Erythropoietin/pharmacology , Gene Expression Regulation , HEK293 Cells , Humans , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nucleoside-Diphosphate Kinase/pharmacology , Nucleoside-Diphosphate Kinase/therapeutic use , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
6.
J Alzheimers Dis ; 39(3): 545-56, 2014.
Article in English | MEDLINE | ID: mdl-24217276

ABSTRACT

The CD147 protein is a ubiquitous multifunctional membrane receptor. Expression of CD147, which is regulated by sterol carrier protein, reportedly modulates amyloid-ß (Aß), the neurotoxic peptide implicated in neuronal degeneration in Alzheimer's disease (AD). Given that high fat/cholesterol is linked to amyloid deposition in AD, we investigated if cholesterol and/or Aß can alter CD147 expression in rat cortical neuronal cultures. Water-soluble cholesterol and Aß42 dose-dependently increased CD147 protein expression, but reduced FL-AßPP protein expression. Cholesterol and Aß42 treatment also increased lactate dehydrogenase release but to varying degrees. Upregulation of CD147 expression was probably mediated by oxidative stress, as H2O2 (3 µM) also induced CD147 protein expression in neuronal cultures. In light of these findings, we investigated if CD147 induction was cytoprotective, a compensatory response to injury, or alternatively, a cell death signal. To this end, we used recombinant adenovirus to overexpress human CD147 (in SH-SY5Y cells and primary cortical neurons), and pre-treated cultures with or without recombinant cyclophilin A (rCYPA) protein, prior to Aß42 exposure. We showed that increased CD147 expression protected against Aß42, only when rCYPA protein was added to neuronal cultures. Together, our findings reveal potentially important relationships between cholesterol loading, CD147 expression, Aß toxicity, and the putative involvement of CYPA protein in neuroprotection in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Basigin/metabolism , Cholesterol/pharmacology , Cyclophilin A/metabolism , Hydrogen Peroxide/pharmacology , Neurons/drug effects , Peptide Fragments/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cyclophilin A/genetics , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Rats , Rats, Sprague-Dawley , Transduction, Genetic
7.
J Neurosci Res ; 92(4): 506-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24375786

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (lrrk2) gene are the leading genetic cause of Parkinson's disease (PD). In characterizing the novel ROC domain mutant A1442P, we compared its steady-state protein levels, propensity to aggregate, and toxicity with the pathogenic R1441C mutant and wild-type (WT) LRRK2. Mutant (R1441C and A1442P) and WT LRRK2 fused to green fluorescent protein (GFP) and FLAG were transiently expressed in HEK293 cells using plasmid constructs. Western analysis and fluorescence microscopy consistently demonstrated lower mutant LRRK2 protein levels compared with WT. A time-course expression study using flow cytometry showed that WT LRRK2 expression increased initially but then plateaued by 72 hr. Conversely, R1441C and A1442P mutant expression attained 85% and 74% of WT levels at 24 hr but fell to 68% and 55% of WT levels by 72 hr, respectively. We found that proteasome inhibition markedly increased mutant LRRK2 to levels approaching those of WT. Taken together, our findings reveal increased intracellular degradation for both mutants. Furthermore, the impact of mutant and WT LRRK2 expression on HEK293 cell viability was assessed under normative and oxidative (hydrogen peroxide) conditions and found not to differ. Expression of WT and mutant LRRK2 protein gave rise to intracellular aggregates of similar appearance and cellular localization. In summary, we provide evidence that the novel A1442P mutant and the previously investigated R1441C pathogenic mutant exhibit increased intracellular degradation, a property reportedly demonstrated for the pathogenic LRRK2 kinase domain mutant I2020T.


Subject(s)
Gene Expression Regulation/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Amino Acids/genetics , Analysis of Variance , Cell Survival , Cysteine Proteinase Inhibitors/pharmacology , Flow Cytometry , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Leupeptins/pharmacology , Time Factors , Transfection
8.
Mol Neurobiol ; 47(2): 821-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23315303

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating and often fatal neurodegenerative disease that affects spinal motor neurons and leads to progressive muscle wasting and paralysis. The survival of motor neuron (SMN) gene is mutated or deleted in most forms of SMA, which results in a critical reduction in SMN protein. Motor neurons appear particularly vulnerable to reduced SMN protein levels. Therefore, understanding the functional role of SMN in protecting motor neurons from degeneration is an essential prerequisite for the design of effective therapies for SMA. To this end, there is increasing evidence indicating a key regulatory antiapoptotic role for the SMN protein that is important in motor neuron survival. The aim of this review is to highlight key findings that support an antiapoptotic role for SMN in modulating cell survival and raise possibilities for new therapeutic approaches.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Survival of Motor Neuron 1 Protein/physiology , Animals , Cell Survival/genetics , Cell Survival/physiology , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/physiology
9.
Neurosci Res ; 73(3): 191-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22561287

ABSTRACT

In this study we have assessed sodium-calcium exchanger (NCX) protein over-expression on cell viability in primary rat cortical neuronal and HEK293 cell cultures when subjected to oxygen-glucose deprivation (OGD). In cortical neuronal cultures, NCX2 and NCX3 over-expression was achieved using adenoviral vectors, and following OGD increased neuronal survival from ≈20% for control vector treated cultures to ≈80% for both NCX isoforms. In addition, we demonstrated that NCX2 and NCX3 over-expression in cortical neuronal cultures enables neurons to maintain intracellular calcium at significantly lower levels than control vector treated cultures when exposed to high (9mM) extracellular calcium challenge. Further assessment of NCX activity during OGD was performed using HEK293 cell lines generated to over-express NCX1, NCX2 or NCX3 isoforms. While it was shown that NCX isoform expression differed considerably in the different HEK293 cell lines, high levels of NCX over-expression was associated with increased resistance to OGD. Taken together, our findings show that high levels of NCX over-expression increases neuronal and HEK293 cell survival following OGD, improves calcium management in neuronal cultures and provides additional support for NCX as a therapeutic target to reduce ischemic brain injury.


Subject(s)
Brain Infarction/metabolism , Glucose/deficiency , Hypoxia-Ischemia, Brain/metabolism , Nerve Degeneration/metabolism , Oxygen/metabolism , Sodium-Calcium Exchanger/biosynthesis , Animals , Brain Infarction/genetics , Brain Infarction/physiopathology , Calcium Signaling/genetics , Female , HEK293 Cells , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Primary Cell Culture , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Rats , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/physiology
10.
J Alzheimers Dis ; 26(4): 593-605, 2011.
Article in English | MEDLINE | ID: mdl-21694447

ABSTRACT

CD147, also known as basigin, EMMPRIN, neurothelin, TCSF, M6, HT7, OX47, or gp42, is a transmembrane glycoprotein of the immunoglobulin super-family. It is expressed in many neuronal and non-neuronal tissues including the hippocampus, pre-frontal cortex thyroid, heart, early erythroid, amygdala, and placenta. This protein is involved in various cellular and biological functions, such as lymphocyte migration and maturation, tissue repair cancer progression, T and B lymphocyte activation, and induction of extracellular matrix metalloproteinase. The CD147 protein interacts with other proteins such as cyclophilin A (CyPA), Cyclophilin B (CyPB), sterol carrier protein (SCP), caveolin-1 and integrins, and can influence amyloid-ß (Aß) peptide levels, a protein that is central to Alzheimer's disease (AD) pathogenesis. Mechanisms by which CD147 regulate Aß levels remain unclear, thus in this review we discuss its involvement in Aß production and clearance and potential mechanisms by which controlling CD147 levels could impact on Aß accumulation and AD pathogenesis.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Basigin/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Basigin/biosynthesis , Basigin/chemistry , Basigin/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cholesterol/pharmacology , Gene Expression Regulation/genetics , Humans
11.
J Neurosci Res ; 85(14): 3089-97, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17663478

ABSTRACT

We previously reported that peroxiredoxin 2 (PRDX2) and Cu/Zn superoxide dismutase 1 (SOD1) proteins are up-regulated in rat primary neuronal cultures following erythropoietin (EPO) preconditioning. In the present study, we have demonstrated that adenovirally mediated overexpression of PRDX2 in cortical neuronal cultures can protect neurons from in vitro ischemia (oxygen-glucose deprivation) and an oxidative insult (cumene hydroperoxide) but not glutamate excitotoxicity. We have also demonstrated that adenovirally mediated overexpression of SOD1 in cortical neuronal cultures protected neurons only against the oxidative insult. Interestingly, we did not detect up-regulation of PRDX2 or SOD1 protein in the rat hippocampus following exposure to either 3 min or 8 min of global cerebral ischemia. Further characterization of PRDX2's neuroprotective mechanisms may aid in the development of a neuroprotective therapy.


Subject(s)
Cerebral Cortex/cytology , Ischemia/prevention & control , Neurons/metabolism , Peroxiredoxins/metabolism , Superoxide Dismutase/metabolism , Adenoviridae/physiology , Analysis of Variance , Animals , Benzene Derivatives/pharmacology , Cell Count/methods , Cells, Cultured , Drug Interactions , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Ischemic Preconditioning/methods , Neurons/drug effects , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Superoxide Dismutase-1 , Time Factors , Transduction, Genetic/methods
12.
Neurobiol Dis ; 25(1): 54-64, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17011206

ABSTRACT

We previously reported that cyclophilin A protein is up-regulated in cortical neuronal cultures following several preconditioning treatments. In the present study, we have demonstrated that adenoviral-mediated over-expression of cyclophilin A in rat cortical neuronal cultures can protect neurons from oxidative stress (induced by cumene hydroperoxide) and in vitro ischemia (induced by oxygen glucose deprivation). We subsequently demonstrated that cultured neurons, but not astrocytes, express the recently identified putative cyclophilin A receptor, CD147 (also called neurothelin, basigin and EMMPRIN), and that administration of purified cyclophilin A protein to neuronal cultures induces a rapid but transient phosphorylation of the extracellular signal-regulated kinase (ERK) 1/2. Furthermore, administration of purified cyclophilin A protein to neuronal cultures protects neurons from oxidative stress and in vitro ischemia. Interestingly, we detected up-regulation of cyclophilin A mRNA, but not protein in the hippocampus following a 3-min period of sublethal global cerebral ischemia in the rat. Despite our in vivo findings, our in vitro data show that cyclophilin A has both intracellular- and extracellular-mediated neuroprotective mechanisms. To this end, we propose cyclophilin A's extracellular-mediated neuroprotection occurs via CD147 receptor signalling, possibly by activation of ERK1/2 pro-survival pathways. Further characterization of cyclophilin A's neuroprotective mechanisms may aid the development of a neuroprotective therapy.


Subject(s)
Basigin/metabolism , Brain Ischemia/pathology , Cyclophilin A/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Neurons/pathology , Oxidative Stress/physiology , Signal Transduction/physiology , Adenoviridae/genetics , Animals , Benzene Derivatives/toxicity , Blotting, Western , Cells, Cultured , Cyclophilin A/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Glucose/deficiency , Hippocampus/cytology , Hippocampus/metabolism , Hypoxia/pathology , Immunohistochemistry , Microscopy, Fluorescence , Oxidants/toxicity , Plasmids/genetics , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/physiology
13.
Brain Res ; 1102(1): 27-38, 2006 Aug 02.
Article in English | MEDLINE | ID: mdl-16806110

ABSTRACT

In order to investigate protein function in rat primary cortical neuronal cultures, we modified an adenoviral vector expression system and assessed the strength and specificity of the cytomegalovirus (CMV), rous sarcoma virus (RSV), and rat and human synapsin 1 (SYN1) promoters to drive DsRed-X expression. We also incorporated the woodchuck post-transcriptional regulatory element (WPRE) and a CMV promoter-enhanced green fluorescent protein (EGFP) reporter cassette. We observed that the RSV promoter activity was strong in neurons and moderate in astrocytes, while the CMV promoter activity was weak-to-moderate in neurons and very strong in astrocytes. The rat and human SYN1 promoters exhibited similar but weak activity in neurons, despite inclusion of the WPRE. We confirmed that the WPRE enhanced RSV promoter-mediated DsRed-X expression in a time-dependent fashion. Interestingly, we observed very weak SYN1-mediated DsRed-X expression in astrocytes and HEK293 cells suggesting incomplete neuronal-restrictive behavior for this promoter. Finally, using our adenoviral expression system, we demonstrated that RSV promoter-mediated Bcl-X(L) overexpression attenuated neuronal death caused by in vitro ischemia and oxidative stress.


Subject(s)
Avian Sarcoma Viruses/genetics , Cytomegalovirus/genetics , Genetic Vectors/physiology , Hepatitis B Virus, Woodchuck/genetics , Neurons/metabolism , Synapsins/metabolism , Animals , Astrocytes/metabolism , Astrocytes/virology , Benzene Derivatives/metabolism , Blotting, Western/methods , Cell Death/physiology , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Gene Expression Regulation, Viral/physiology , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry/methods , Neurons/virology , Promoter Regions, Genetic/physiology , Rats , Regulatory Sequences, Nucleic Acid , Synapsins/genetics , Time Factors , Transgenes
14.
J Neurosci Res ; 83(4): 584-93, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16435392

ABSTRACT

In this study we confirmed the presence of the erythropoietin (EPO) receptor on both cultured cortical neurons and PC12 cells and showed that EPO can induce changes in p38, ERK, and JNK signaling molecules in these cells. We induced EPO preconditioning in cortical neuronal cultures that protected neurons from a subsequent in vitro ischemic insult (transient oxygen-glucose deprivation). To investigate downstream changes in protein expression in EPO-preconditioned cortical neuronal cultures, we used two-dimensional gel electrophoresis. Overall, EPO preconditioning resulted in protein up-regulation, and, from 84 of the most differentially expressed proteins selected for identification, the proteins or tentative proteins were identified in 57 cases, representing 40 different proteins. Different protein spots representing the same or closely related protein(s) occurred for 13 of the identified proteins and are likely to represent posttranslational modifications or proteolytic fragments of the protein. Two proteins (78-kD glucose-regulated protein and tropomyosin, fibroblast isoform 1) were detected in control neuronal cultures, but not following EPO preconditioning treatment, whereas one protein (40S ribosomal protein SA) was detected only following EPO preconditioning. Most of the other proteins identified had not previously been associated with EPO preconditioning and will aid in the understanding of EPO's neuroprotective response and possibly the development of new therapeutic interventions to inhibit neuronal death in acute and chronic neurodegenerative diseases.


Subject(s)
Erythropoietin/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/drug effects , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Electrophoresis, Gel, Two-Dimensional , Image Processing, Computer-Assisted , Immunoblotting , Immunohistochemistry , Ischemia/pathology , Neurons/metabolism , Neuroprotective Agents/pharmacology , PC12 Cells , Proteomics , Rats , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stimulation, Chemical , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL
...