Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cytotherapy ; 23(9): 852-860, 2021 09.
Article in English | MEDLINE | ID: mdl-33941482

ABSTRACT

BACKGROUND AIMS: Next-generation immune cell therapy products will require complex modifications using engineering technologies that can maintain high levels of cell functionality. Non-viral engineering methods have the potential to address limitations associated with viral vectors. However, while electroporation is the most widely used non-viral modality, concerns about its effects on cell functionality have led to the exploration of alternative approaches. Here the authors have examined the suitability of the Solupore non-viral delivery system for engineering primary human T cells for cell therapy applications. METHODS: The Solupore system was used to deliver messenger RNA (mRNA) and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) guide RNA ribonucleoprotein (RNP) cargos to T cells, and efficiency was measured by flow cytometry. Cell perturbation was assessed by immune gene expression profiling, including an electroporation comparator. In vitro and in vivo cytotoxicity of chimeric antigen receptor (CAR) T cells generated using the Solupore system was evaluated using a real-time cellular impedance assay and a Raji-luciferase mouse tumor model, respectively. RESULTS: Efficient transfection was demonstrated through delivery of mRNA and CRISPR CAS9 RNP cargos individually, simultaneously and sequentially using the Solupore system while consistently maintaining high levels of cell viability. Gene expression profiling revealed minimal alteration in immune gene expression, demonstrating the low level of perturbation experienced by the cells during this transfection process. By contrast, electroporation resulted in substantial changes in immune gene expression in T cells. CAR T cells generated using the Solupore system exhibited efficient cytotoxicity against target cancer cells in vitro and in vivo. CONCLUSIONS: The Solupore system is a non-viral means of simply, rapidly and efficiently delivering cargos to primary human immune cells with retention of high cell viability and functionality.


Subject(s)
Genetic Vectors , T-Lymphocytes , Animals , Cell- and Tissue-Based Therapy , Electroporation , Humans , Mice , Transfection
2.
PLoS One ; 12(3): e0174779, 2017.
Article in English | MEDLINE | ID: mdl-28358921

ABSTRACT

Despite advances in intracellular delivery technologies, efficient methods are still required that are vector-free, can address a wide range of cargo types and can be applied to cells that are difficult to transfect whilst maintaining cell viability. We have developed a novel vector-free method that uses reversible permeabilization to achieve rapid intracellular delivery of cargos with varying composition, properties and size. A permeabilizing delivery solution was developed that contains a low level of ethanol as the permeabilizing agent. Reversal of cell permeabilization is achieved by temporally and volumetrically controlling the contact of the target cells with this solution. Cells are seeded in conventional multi-well plates. Following removal of the supernatant, the cargo is mixed with the delivery solution and applied directly to the cells using an atomizer. After a short incubation period, permeabilization is halted by incubating the cells in a phosphate buffer saline solution that dilutes the ethanol and is non-toxic to the permeabilized cells. Normal culture medium is then added. The procedure lasts less than 5 min. With this method, proteins, mRNA, plasmid DNA and other molecules have been delivered to a variety of cell types, including primary cells, with low toxicity and cargo functionality has been confirmed in proof-of-principle studies. Co-delivery of different cargo types has also been demonstrated. Importantly, delivery occurs by diffusion directly into the cytoplasm in an endocytic-independent manner. Unlike some other vector-free methods, adherent cells are addressed in situ without the need for detachment from their substratum. The method has also been adapted to address suspension cells. This delivery method is gentle yet highly reproducible, compatible with high throughput and automated cell-based assays and has the potential to enable a broad range of research, drug discovery and clinical applications.


Subject(s)
Cell Membrane Permeability/physiology , A549 Cells , Cell Membrane Permeability/genetics , Drug Delivery Systems , Electroporation , Flow Cytometry , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics
3.
J Mol Endocrinol ; 56(3): 261-71, 2016 04.
Article in English | MEDLINE | ID: mdl-26759391

ABSTRACT

Metformin is the main drug of choice for treating type 2 diabetes, yet the therapeutic regimens and side effects of the compound are all undesirable and can lead to reduced compliance. The aim of this study was to elucidate the mechanism of action of two novel compounds which improved glucose handling and weight gain in mice on a high-fat diet. Wildtype C57Bl/6 male mice were fed on a high-fat diet and treated with novel, anti-diabetic compounds. Both compounds restored the glucose handling ability of these mice. At a cellular level, these compounds achieve this by inhibiting complex I activity in mitochondria, leading to AMP-activated protein kinase activation and subsequent increased glucose uptake by the cells, as measured in the mouse C2C12 muscle cell line. Based on the inhibition of NADH dehydrogenase (IC50 27µmolL(-1)), one of these compounds is close to a thousand fold more potent than metformin. There are no indications of off target effects. The compounds have the potential to have a greater anti-diabetic effect at a lower dose than metformin and may represent a new anti-diabetic compound class. The mechanism of action appears not to be as an insulin sensitizer but rather as an insulin substitute.


Subject(s)
Diet, High-Fat , Electron Transport Complex I/antagonists & inhibitors , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Piperazines/pharmacology , Thiophenes/pharmacology , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Hypoglycemic Agents/chemistry , Male , Mice , NAD/metabolism , Oxygen Consumption , Rats
4.
Proteomics ; 16(1): 70-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26573124

ABSTRACT

Elevated serum retinol-binding protein (RBP) concentration has been implicated in the development of insulin resistance and type 2 diabetes. Two series of small molecules have been designed to lower serum levels by reducing secretion of the transthyretin-RBP complex from the liver and enhancing RBP clearance through the kidney. These small molecules were seen to improve glucose and insulin tolerance tests and to reduce body weight gain in mice rendered diabetic through a high fat diet. A proteomics study was conducted to better understand the effects of these compounds in muscle cells, muscle being the primary site for energy expenditure. One lead compound, RTC-15, is seen to have a significant effect on proteins involved in fat and glucose metabolism. This could indicate that the compound is having a direct effect on muscle tissue to improve energy homeostasis as well as a whole body effect on circulating RBP levels. This newly characterized group of antidiabetic compounds may prove useful in the treatment and prevention of insulin resistance and obesity.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Muscle Cells/drug effects , Protein Interaction Maps/drug effects , Proteome/metabolism , Animals , Cell Line , Glucose/metabolism , Hypoglycemic Agents/chemistry , Insulin Resistance , Mice , Muscle Cells/metabolism , Retinol-Binding Proteins, Plasma/metabolism
5.
Diabetes Metab Res Rev ; 32(4): 379-90, 2016 May.
Article in English | MEDLINE | ID: mdl-26556762

ABSTRACT

BACKGROUND: Retinol binding protein (RBP) and its membrane receptor, STRA6, are vital for the management of vitamin A in the body. Recently, elevated serum RBP levels have been implicated as a contributing factor to the development of insulin resistance and type 2 diabetes. However, conflicting opinions exist as to how these increased levels can cause insulin resistance. METHODS: In order to better understand the influences of RBP, a proteomic study was devised to determine the direct effect of RBP on a mouse muscle cell line, because the muscle is the principal site of insulin induced glucose uptake. C2C12 cells were treated with RBP for 16 h and the proteome analysed for alterations in protein abundance and phosphorylation by 2-DE. RESULTS: A number of changes were observed in response to retinol binding protein treatment, of which the most interesting were decreased levels of the phosphatase, protein phosphatase 1 ß. This phosphatase is responsible for regulating glycogen synthase and glycogen phosphorylase, the rate-limiting enzymes involved in glycogen storage and utilization. Retinol binding protein treatment resulted in increased phosphorylation and inhibition of glycogen synthase, with detrimental effects on insulin stimulated glycogen production in these cells. CONCLUSION: The results indicate that RBP may have a negative effect on energy storage in the cell and could contribute to the development of insulin resistance in muscle tissue. Understanding how retinol binding protein influences insulin resistance may reveal novel strategies to target this disease.


Subject(s)
Biomarkers/metabolism , Muscle Cells/metabolism , Proteome/analysis , Retinol-Binding Proteins/pharmacology , Animals , Cells, Cultured , Chromatography, Liquid , Immunoblotting , Immunoprecipitation , Mice , Muscle Cells/drug effects , Tandem Mass Spectrometry
6.
PLoS One ; 10(3): e0122293, 2015.
Article in English | MEDLINE | ID: mdl-25816144

ABSTRACT

STRA6 is a plasma membrane protein that mediates the transport of vitamin A, or retinol, from plasma retinol binding protein (RBP) into the cell. Mutations in human STRA6 are associated with Matthew-Wood syndrome, which is characterized by severe developmental defects. Despite the obvious importance of this protein to human health, little is known about its structure and mechanism of action. To overcome the difficulties frequently encountered with the production of membrane proteins for structural determination, STRA6 has been expressed in Pichia pastoris as a fusion to green fluorescent protein (GFP), a strategy which has been a critical first step in solving the crystal structures of several membrane proteins. STRA6-GFP was correctly targeted to the cell surface where it bound RBP. Here we report the large-scale expression, purification and characterisation of STRA6-GFP. One litre of culture, corresponding to 175 g cells, yielded about 1.5 mg of pure protein. The interaction between purified STRA6 and its ligand RBP was studied by surface plasmon resonance-based binding analysis. The interaction between STRA6 and RBP was not retinol-dependent and the binding data were consistent with a transient interaction of 1 mole RBP/mole STRA6.


Subject(s)
Membrane Proteins/biosynthesis , Membrane Proteins/isolation & purification , Pichia/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/genetics , Pichia/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Surface Plasmon Resonance
7.
Neuroimmunomodulation ; 16(6): 400-10, 2009.
Article in English | MEDLINE | ID: mdl-19609089

ABSTRACT

BACKGROUND: A common change that occurs with age in the central nervous system is an increase in microglial-associated inflammation. This is usually coupled with an increase in the concentration of the inflammatory cytokine interleukin-1beta (IL-1beta) in the hippocampus and an inhibition in long-term potentiation. OBJECTIVES: To assess the effects of a novel preparation of phospholipid nanoparticles incorporating phosphatidylglycerol, VP025, on inflammatory changes in hippocampus of aged and lipopolysaccharide (LPS)-treated rats. METHODS/RESULTS: We report that a possible initial target cell of the putative anti-inflammatory actions of VP025 may be macrophages, as VP025 is engulfed by, and has the capacity to alter the activity of, these cells. VP025 reversed the increase in IFN-gamma concentration in supernatant taken from peritoneal macrophages harvested from LPS-treated rats. In addition, markers of microglial activity, major histocompatibility complex class II (MHC II) mRNA expression, CD40 expression and IL-1beta concentration were increased, and CD200 expression was reduced, in the hippocampus of these rats. VP025 reversed changes in CD40, IL-1beta and CD200 in aged rats, and also restored long-term potentiation in aged and LPS-treated rats. CONCLUSIONS: We conclude that VP025 has the ability to modulate the activity of macrophage, microglia and neurons in response to stressors such as ageing and LPS treatment.


Subject(s)
Aging/physiology , Anti-Inflammatory Agents/pharmacology , Encephalitis/drug therapy , Gliosis/drug therapy , Microglia/drug effects , Phosphatidylglycerols/pharmacology , Phospholipids/pharmacology , Adult , Animals , Anti-Inflammatory Agents/chemistry , Encephalitis/immunology , Encephalitis/physiopathology , Gliosis/chemically induced , Gliosis/physiopathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Immunomodulation/drug effects , Immunomodulation/physiology , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Macrophages/drug effects , Macrophages/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Memory Disorders/physiopathology , Microglia/physiology , Nanoparticles/chemistry , Perforant Pathway/drug effects , Perforant Pathway/metabolism , Perforant Pathway/physiopathology , Phagocytosis/drug effects , Phagocytosis/physiology , Phosphatidylglycerols/chemistry , Phospholipids/chemistry , Rats , Rats, Wistar , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
8.
Exp Gerontol ; 44(4): 300-4, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19121379

ABSTRACT

Amyloid-beta (Abeta) peptides, the primary component of the amyloid plaques in Alzheimer's disease (AD), exert profound effects on neurons in vitro and negatively impact on neuronal function in vivo. One of the consequences of increased Abeta in the brain, either as a result of overexpression of the precursor amyloid precursor protein in transgenic mice, or injection into the brain is a decrease in one form of synaptic plasticity, long-term potentiation (LTP) in the hippocampus. Here we investigated the effect of infusion of Abeta for 28 days on LTP in dentate gyrus of rats and demonstrate that it was profoundly decreased compared with control-treated rats. We show that this effect is accompanied by increased activity of caspase 3, which is an indicator of cell stress. Significantly these changes were attenuated in animals which were pretreated with particles incorporating phosphatidylglycerol (VP025) and the evidence indicated that even when treatment was given 2 weeks after the start of the Abeta infusion, VP025 was capable of attenuating Abeta-induced changes. The evidence suggests that activation of caspase 3 was mediated by an Abeta-induced increase in sphingomyelinase, with the subsequent production of ceramide which is known to have a detrimental effect on neuronal function.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Caspase 3/drug effects , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Amyloid beta-Peptides/metabolism , Animals , Caspase 3/metabolism , Electric Stimulation , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Rats , Rats, Wistar
9.
J Biol Chem ; 280(10): 9354-62, 2005 Mar 11.
Article in English | MEDLINE | ID: mdl-15615726

ABSTRACT

It is well documented that long term potentiation (LTP) is impaired in the hippocampus of the aged animal. Among the changes that contribute to this impairment is an increase in hippocampal concentration of the pro-inflammatory cytokine interleukin-1beta (IL-1beta), and increased IL-1beta-induced signaling. In this study we investigated the possibility that these changes were a consequence of decreased concentration of the anti-inflammatory cytokine, IL-4, and decreased IL-4-stimulated signaling. We report that functional IL-4 receptors are expressed on granule cells of the dentate gyrus and that receptor activation results in phosphorylation of JAK1 and STAT6. Hippocampal IL-4 concentration was decreased with age, and this was accompanied by a decrease in phosphorylation of JAK1 and STAT6. The evidence indicates that IL-4 modulates expression of IL-1beta mRNA and protein and that it attenuates IL-1beta-induced impairment of LTP and phosphorylation of JNK and c-Jun. We argued that, if a decrease in hippocampal IL-4 concentration significantly contributed to the age-related impairment in LTP, then restoration of IL-4 should restore LTP. To test this, we treated rats with VP015 (phospholipid microparticles-incorporating phosphatidylserine), which increases IL-4 concentration in hippocampus. The data indicate that the VP015-induced increase in IL-4 concentration in hippocampus of aged rats and lipopolysaccharide (LPS)-treated rats was accompanied by a reversal of the age-related and LPS-induced impairment in LTP in perforant path granule cell synapses. We propose that interplay between pro-inflammatory and anti-inflammatory responses impact significantly on synaptic function in the hippocampus of the aged rat.


Subject(s)
Hippocampus/physiopathology , Inflammation/prevention & control , Inflammation/physiopathology , Interleukin-4/physiology , Aging , Animals , Anti-Inflammatory Agents , DNA Primers , Dentate Gyrus/drug effects , Dentate Gyrus/physiopathology , Hippocampus/growth & development , Interleukin-1/genetics , Interleukin-4/genetics , Long-Term Potentiation , Male , Rats , Rats, Wistar , Receptors, Interleukin-1/genetics , Receptors, Interleukin-4/physiology , Signal Transduction
10.
J Neurochem ; 91(1): 20-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15379883

ABSTRACT

Eicosapentaenoic acid (EPA) protects hippocampus from age-related and irradiation-induced changes that lead to impairment in synaptic function; the evidence suggests that this is due to its anti-inflammatory effects, specifically preventing changes induced by the proinflammatory cytokine, interleukin-1beta (IL-1beta). In this study, we have investigated the possibility that EPA may prevent the effects of lipopolysaccharide (LPS) administration, which have been shown to lead to deterioration of synaptic function in rat hippocampus. The data indicate that treatment of hippocampal neurones with EPA abrogated the LPS-induced increases in phosphorylation of the mitogen-activated protein kinase, c-Jun N-terminal kinase (JNK), the transcription factor, c-Jun and the mitochondrial protein, Bcl-2. In parallel, we report that intraperitoneal administration of LPS to adult rats increases phosphorylation of JNK, c-Jun and Bcl-2 in hippocampal tissue and that these changes are coupled with increased IL-1beta concentration. Treatment of rats with EPA abrogates these effects and also blocks the LPS-induced impairment in long-term potentiation in perforant path-granule cell synapses that accompanies these changes. We propose that the neuroprotective effect of EPA may be dependent on its ability to inhibit the downstream consequences of JNK activation.


Subject(s)
Brain Diseases/prevention & control , Eicosapentaenoic Acid/therapeutic use , Hippocampus/physiology , Neuroprotective Agents/therapeutic use , Animals , Animals, Newborn , Blotting, Western/methods , Brain Diseases/chemically induced , Cells, Cultured , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Drug Interactions , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/injuries , Interleukin-1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides , Long-Term Potentiation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neural Inhibition/drug effects , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Wistar , Time Factors , bcl-2-Associated X Protein
11.
J Biol Chem ; 278(51): 51075-84, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14525971

ABSTRACT

Among the many reported effects of irradiation in cells is activation of the stress-activated protein kinase, c-Jun N-terminal kinase (JNK), which has been shown to result in apoptotic cell death. The trigger that leads to JNK activation has not been identified, although, in rat hippocampus at least, irradiation-induced apoptosis has been coupled with increased accumulation of reactive oxygen species (ROS). Significantly, irradiation-induced changes in hippocampus are abrogated by treatment of rats with the polyunsaturated fatty acid, eicosapentaenoic acid (EPA). A close coupling between ROS accumulation and concentration of the pro-inflammatory cytokine, interleukin-1 beta (IL-1 beta) in hippocampus has been reported, and the evidence suggests that IL-1 beta may be responsible for the enhanced ROS production. Here we set out to assess the possibility that whole body gamma-irradiation increases IL-1 beta concentration in hippocampus and to investigate the consequences of such a change. We present evidence that reveals that the irradiation-induced increase in IL-1 beta concentration in hippocampus is accompanied by increased expression of IL-1 type I receptor and IL-1 accessory protein and increased activation of IL-1 receptor-activated kinase. These changes, which were coupled with increased activation of JNK and evidence of apoptotic cell death, were absent in hippocampus of rats that received EPA treatment. Significantly, EPA treatment enhanced hippocampal IL-10 concentration that was inversely correlated with IL-1 beta concentration. The data are consistent with the idea that EPA exerts anti-inflammatory and neuroprotective effects in the central nervous system.


Subject(s)
Gamma Rays , Hippocampus/metabolism , Interleukin-1/radiation effects , Signal Transduction/radiation effects , Animals , Apoptosis , Eicosapentaenoic Acid , Fatty Acids, Unsaturated/pharmacology , Hippocampus/cytology , Interleukin-10/analysis , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/metabolism , Protective Agents/pharmacology , Rats , Rats, Wistar , Up-Regulation , Whole-Body Irradiation
12.
J Biol Chem ; 277(37): 34239-46, 2002 Sep 13.
Article in English | MEDLINE | ID: mdl-12091394

ABSTRACT

Among the several changes that occur in the aged brain is an increase in the concentration of the proinflammatory cytokine interleukin-1beta that is coupled with a deterioration in cell function. This study investigated the possibility that treatment with the polyunsaturated fatty acid eicosapentaenoic acid might prevent interleukin-1beta-induced deterioration in neuronal function. Assessment of four markers of apoptotic cell death, cytochrome c translocation, caspase-3 activation, poly(ADP-ribose) polymerase cleavage, and terminal dUTP nick-end staining, revealed an age-related increase in each of these measures, and the evidence presented indicates that treatment of aged rats with eicosapentaenoate reversed these changes as well as the accompanying increases in interleukin-1beta concentration and p38 activation. The data are consistent with the idea that activation of p38 plays a significant role in inducing the changes described since interleukin-1beta-induced activation of cytochrome c translocation and caspase-3 activation in cortical tissue in vitro were reversed by the p38 inhibitor SB203580. The age-related increases in interleukin-1beta concentration and p38 activation in cortex were mirrored by similar changes in hippocampus. These changes were coupled with an age-related deficit in long term potentiation in perforant path-granule cell synapses, while eicosapentaenoate treatment was associated with reversal of age-related changes in interleukin-1beta and p38 and with restoration of long term potentiation.


Subject(s)
Aging/pathology , Apoptosis/drug effects , Arachidonic Acids/pharmacology , Brain/pathology , Interleukin-1/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Animals , Enzyme Activation/drug effects , In Situ Nick-End Labeling , Long-Term Potentiation/drug effects , Male , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases
13.
J Biol Chem ; 277(23): 20804-11, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-11912218

ABSTRACT

Exposure to irradiation leads to detrimental changes in several cell types. In this study we assessed the changes induced in hippocampus by exposure of rats to whole body irradiation; the findings reveal that irradiation leads to apoptotic cell death in hippocampus, and as a consequence, long term potentiation in perforant path-granule cell synapses is markedly impaired. The evidence is consistent with the view that irradiation induced an increase in reactive oxygen species and that this leads to stimulation of the stress-activated protein kinase, JNK, and activation of the transcription factor, c-Jun. Consequent upon activation of JNK, a cascade of cell signaling events was stimulated that ultimately resulted in apoptosis, as suggested by parallel increases in cytochrome c translocation, caspase-3 activation, poly(ADP-ribose) polymerase cleavage, and terminal dUTP nick-end labeling staining. Treatment of rats with eicosapentaenoic acid inhibited the irradiation-induced increase in reactive oxygen species production and the subsequent cellular signaling events, suggesting that oxidative stress triggered apoptotic cell death in the hippocampus of rats exposed to irradiation. Significantly, when the compromise in cell viability induced by irradiation was prevented by eicosapentaenoic acid, long term potentiation was sustained in a manner similar to that in the sham-treated control group.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Hippocampus/radiation effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Cytochrome c Group , Gamma Rays , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases , Long-Term Potentiation/drug effects , Long-Term Potentiation/radiation effects , Male , Mitogen-Activated Protein Kinases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...