Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters








Publication year range
1.
Nurse Educ Today ; 57: 82-87, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28753518

ABSTRACT

Previous studies suggest that theory-practice gap has remained to be a formidable task and a challenge to the nursing profession. While efforts to understand the nature and dynamics of theory-practice gap have been undertaken across the globe, a dearth in literature exists in the context of a developing country like the Philippines. Seemingly, no research has ventured yet to explore the theory-practice gap experiences of Filipino student nurses. Hence, the major intent of this qualitative investigation is to describe and capture how theory-practice gap is viewed by a select group of senior nursing students (n=10) in a comprehensive university in the Philippines. From the thickness and richness of the descriptions of the field text gathered in this study, an interesting conceptualization labeled as the Theory-Practice Gap Deficit Triad which consists of (a) structural, (b) pedagogical, and (c) relational deficits was emerged. Interestingly, the said model describes a clear intersection of the various concerns and dilemmas encountered by student nurses in their clinical exposures. Findings of this paper are valuable inputs to nursing educators, practitioners, and administrators in initiating realignment efforts geared toward developing nurses whose knowledge, skills and attitudes are responsive to the ever changing professional practice landscape.


Subject(s)
Attitude of Health Personnel , Clinical Competence , Education, Nursing, Baccalaureate , Students, Nursing/psychology , Faculty, Nursing , Humans , Philippines , Professional Practice Gaps , Qualitative Research
2.
Nurse Educ Today ; 55: 122-127, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28575709

ABSTRACT

The role of clinical instructor in student nurses' preparation for the professional nursing practice cannot be underestimated. The extent to which such role is achieved depends highly on the instructors' ability to realize the desired qualities expected of them. While a number of empirical studies have qualitatively explored the attributes of an effective clinical instructor, no attempt has ventured yet on the power of experimental vignettes for conjoint analysis in explicating the preferences of a select group of Filipino student nurses relative to their clinical instructors' attributes. Junior and senior nursing students (n=227), recruited from one of the comprehensive universities in the Philippines, were asked to sort out orthogonal cards generated by Sawtooth Software. As shown, the full-profile conjoint analysis was considerably fit for this study: Pearson's R=0.988, (p<0.05) and Kendall's t=0.944, (p<0.05). Results indicated that the student nurses are one in terms of their most preferred clinical instructor attribute, which was clinical teaching capacity (38.14%) followed by interpersonal relationship and caring behavior (33.17%). In regard to the clinical teaching capability, a clinical instructor who parallels clinical teaching skills with the students' understanding and experience (0.089) was the highest part-worth. As for the interpersonal relationship and caring behavior, the highest part-worth was a clinical instructor who respects a student nurse as an individual and cares about him/her as a person (0.114). Findings of this study can be a basis for clinical instructors as to which qualities to cultivate best to facilitate a first-rate clinical nursing instruction. Likewise, the results of this study can inform current practices of clinical instructors by making them aware of how they can nurture a pedagogical approach consistent with the student nurses' preferences.


Subject(s)
Clinical Competence/standards , Empathy , Faculty, Nursing/psychology , Interpersonal Relations , Students, Nursing/psychology , Achievement , Education, Nursing, Baccalaureate , Humans , Philippines
3.
Neuroscience ; 107(4): 571-84, 2001.
Article in English | MEDLINE | ID: mdl-11720781

ABSTRACT

In vitro ischemic preconditioning induced by subjecting rat cortical cultures to nonlethal oxygen-glucose deprivation protects against a subsequent exposure to otherwise lethal oxygen-glucose deprivation. We provide evidence that attenuation of the postsynaptic N-methyl-D-aspartate (NMDA) receptor- and Ca(2+)-dependent neurotoxicity underlies oxygen-glucose deprivation tolerance. It is demonstrated that extended tolerance to otherwise lethal NMDA or oxygen-glucose deprivation can be induced by either of their sublethal forms of preconditioning. These four pathways are linked, since NMDA receptor blockade during preconditioning by oxygen-glucose deprivation eliminates tolerance. These results suggest that NMDA tolerance, induced by nonlethal activation of these receptors during oxygen-glucose deprivation preconditioning, underlies oxygen-glucose deprivation tolerance. Several neurotoxic downstream Ca(2+)-dependent signaling events specifically linked to NMDA receptor activation are attenuated during otherwise lethal oxygen-glucose deprivation in preconditioned cultures. Specifically, calpain activation, as well as degradation of microtubule-associated protein-2 and postsynaptic density-95, are attenuated 2 h following otherwise lethal NMDA treatment alone or oxygen-glucose deprivation in preconditioned cultures. Formation of microtubule-associated protein-2-labeled dendritic varicosities is also attenuated in preconditioned cultures within 1 h of lethal oxygen-glucose deprivation or NMDA application. Intracellular Ca(2+) levels, measured using the high- or low-affinity dyes Fluo-4 (K(d) approximately equal 345 nM) or Fluo-4FF (K(d) approximately equal 9.7 microM) respectively, are markedly attenuated during lethal oxygen-glucose deprivation in preconditioned cultures.Collectively, the results suggest the attenuation of the postsynaptic NMDA-mediated component of otherwise lethal oxygen-glucose deprivation through the suppression of Ca(2+)-dependent neurotoxic signaling, a mechanism that is initially induced by transient nonlethal activation of this receptor during ischemic preconditioning.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Glucose/pharmacology , Ischemic Preconditioning , N-Methylaspartate/toxicity , Neurons/drug effects , Oxygen/pharmacology , Aniline Compounds , Animals , Biomarkers , Brain Ischemia/physiopathology , Calcium/metabolism , Calpain/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Female , Fluorescent Antibody Technique , Fluorescent Dyes , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Pregnancy , Rats , Xanthenes
4.
Brain Res ; 900(1): 26-37, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11325343

ABSTRACT

During normal development of the central nervous system there is expression of cyclins that regulate the progression of cells through various stages of mitosis. Cyclins have also been implicated in neuronal degeneration and apoptosis in adult brain, especially cyclin D1 as it is permissive for the transition from growth phase to synthesis phase in mitotic cell division. There is controversy as to whether cyclin D1 expression increases in both in vitro and in vivo models of cerebral ischemia. In this study we use immunohistochemistry and Western blot analysis to characterize cyclin D1 expression in an in vivo rat global model of cerebral ischemia to address the hypothesis that cyclin D1 alterations are involved in ischemic neuronal death. Although there was no change in cyclin D1 expression in either the vulnerable CA1 or resistant CA3 regions of the hippocampus prior to neuronal cell death (<3 days reperfusion), concomitant with the death of CA1 neurons and the loss of cyclin D1 in these cells, there was an increase in non-neuronal cyclin D1 positive cells. Some of the non-neuronal cyclin D1 expressing cells were identified to be activated microglia. In contrast to the cytoplasmic expression of cyclin D1 in neurons, the cyclin D1 expression in the microglia and other non-neuronal cells in CA1 was both nuclear and cytosolic. This study suggests that cyclin D1 does not play a role in the death of vulnerable CA1 neurons in global ischemia.


Subject(s)
Brain Ischemia/metabolism , Cyclin D1/biosynthesis , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Animals , Apoptosis , Biomarkers , Blotting, Western , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Cycle , Cyclin D1/genetics , Dentate Gyrus/pathology , Hippocampus/pathology , Male , Microscopy, Confocal , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Neurons/pathology , Pyramidal Cells/pathology , Rats , Rats, Wistar
5.
Acta Neurochir Suppl ; 76: 47-53, 2000.
Article in English | MEDLINE | ID: mdl-11450070

ABSTRACT

Brain inflammation has been implicated in the development of brain edema and secondary brain damage in ischemia and trauma. Mechanisms involved in leukocyte infiltration across the blood-brain barrier are still unknown. In this study, we show that human cere-bromicrovascular endothelial cells (HCEC) subjected to a 4 h in vitro ischemia (hypoxia + glucose deprivation) followed by a 4-24 h recovery express elevated levels of ICAM-1, IL-8, and MCP-1 mRNAs (semi-quantitative RT-PCR) and secrete increased amounts of the immunoreactive chemokines IL-8 and MCP-1 (ELISA). The ischemia-induced expression of ICAM-1 in HCEC, and the expression/release of IL-8 and MCP-1 in HCEC were abolished by the non-steroid anti-inflammatory drug, indomethacin (100-300 microM). The immunosuppressant cyclosporin A (50 microM) partially reduced the ischemia-stimulated IL-8 and MCP-1 secretion by HCEC. Both indomethacin and cyclosporin A also inhibited the ischemia-induced neutrophil chemotaxis elicited by HCEC media. The study indicates that in vitro ischemia augments the expression of adhesion molecules and leukocyte chemoattractants at the site of the BBB. This ischemic pro-inflammatory activation of HCEC may constitute a key event in initiating post-ischemic inflammation, and it can be suppressed by the anti-inflammatory drugs, indomethacin and cyclosporin A.


Subject(s)
Brain Edema/immunology , Brain Ischemia/immunology , Chemokines/antagonists & inhibitors , Cyclosporine/pharmacology , Endothelium, Vascular/drug effects , Indomethacin/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Chemokines/metabolism , Endothelium, Vascular/immunology , Humans , Leukocytes/drug effects , Leukocytes/immunology
6.
J Neuroimmunol ; 101(2): 148-60, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10580798

ABSTRACT

Leukocyte infiltration into the brain has been implicated in the development of ischemic brain damage. In this study, simulated in vitro ischemia/reperfusion and IL-1beta were found to up-regulate both the expression of intercellular adhesion molecule- (ICAM-1) in cultured human cerebromicrovascular endothelial cells (HCEC) and the adhesion of allogenic neutrophils to HCEC. Both HCEC and human fetal astrocytes (FHAS) also responded to IL-1beta and to in vitro ischemia/reperfusion by a pronounced up-regulation of IL-8 and MCP-1 mRNA and by increased release of IL-8 and MCP-1 in cell culture media. FHAS were found to release 30-times higher levels of MCP-1 than HCEC under both basal and ischemic conditions. However, 100 u/ml IL-1beta induced greater stimulation of both IL-8 and MCP-1 secretion in HCEC (50 and 20 times above controls, respectively) than in FHAS (three and two times above controls, respectively). IL-8 was the principal neutrophil chemoattractant released from IL-1beta-treated HCEC, since IL-8 antibody completely inhibited neutrophil chemotaxis enticed by HCEC media. However, the IL-8 antibody neutralized only 50% of IL-1beta-stimulated neutrophil chemoattractants released from FHAS, and 40%-60% of ischemia-stimulated chemotactic activity released by either HCEC or FHAS. These results suggest that simulated in vitro ischemia, in addition to IL-8 and MCP-1, stimulates secretion of other bioactive chemokines from HCEC and FHAS.


Subject(s)
Astrocytes/metabolism , Brain Ischemia/immunology , Brain/blood supply , Chemokines/biosynthesis , Endothelium, Vascular/metabolism , Blood-Brain Barrier , Brain Ischemia/metabolism , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemotaxis, Leukocyte , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-1/pharmacology , Interleukin-8/biosynthesis , Neutrophils/immunology
7.
Brain Res ; 842(2): 376-83, 1999 Sep 25.
Article in English | MEDLINE | ID: mdl-10526133

ABSTRACT

Recent evidence suggests that apoptosis in post-mitotic neurons involves an aborted attempt of cells to re-enter the cell cycle and it is characterized by increased expression of cyclins, such as cyclin D1, prior to death. Cyclin D1 increases to permit transition from growth phase (G0/G1) to synthesis phase (S) during normal development but there is controversy as to which of the cyclins are activated prior to apoptotic cell death. We looked at the expression of cyclin D1 in cortical neuronal cultures treated with either staurosporine to produce apoptotic death, or with glutamate, to produce a non-apoptotic death. Cyclin D1 immunoreactivity was observed in the cytoplasm and nucleus of virtually all neurons under control conditions. Following the addition of either staurosporine or glutamate, cyclin D1 immunoreactivity did not change within 4 h. The cyclin D1 immunoreactivity was lost by 6 h with the appearance of either staurosporine-induced fragmented nuclei or glutamate-induced pyknotic nuclei. These immunocytochemical observations were confirmed with immunoblot analysis. Therefore, cyclin D1 is not a reliable indicator of apoptosis in cortical neuronal cultures and should not be used as an indicator of apoptotic cell death.


Subject(s)
Apoptosis , Cell Cycle/physiology , Cerebral Cortex/cytology , Cyclin D1/metabolism , Models, Neurological , Nerve Degeneration , Neurons/cytology , Neurons/physiology , Animals , Cells, Cultured , Cerebral Cortex/pathology , Cyclin D1/analysis , Fetus , G1 Phase , Glutamic Acid/pharmacology , Kinetics , Necrosis , Neurons/drug effects , Rats , Resting Phase, Cell Cycle , S Phase , Staurosporine/pharmacology
8.
Neurol Res ; 21(5): 524-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10439436

ABSTRACT

The low-affinity use-dependent N-methyl-D-aspartate (NMDA) receptor antagonist AR-R15896AR is neuroprotective in primary rat cortical cultures exposed to toxic concentrations of NMDA and reduces the magnitude of NMDA-triggered increases in [Ca2+]i. Here we show using fluorescence staining and measurements of microtubule-associated protein-2 (MAP2) levels, that AR-R15896AR inhibits the NMDA-induced loss of MAP2 that occurs within 2 min following NMDA exposure. Understanding the multiple, Ca(2+)-triggered intracellular events that occur following NMDA receptor stimulation is important to the development of safe and effective neuroprotective agents.


Subject(s)
Anticonvulsants/pharmacology , Brain Chemistry/drug effects , Calcium Signaling/drug effects , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Pyridines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cells, Cultured , Cerebral Cortex/cytology , Drug Evaluation, Preclinical , Ionomycin/pharmacology , Ionophores/pharmacology , Microscopy, Fluorescence , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley
9.
Neuroreport ; 9(7): 1287-90, 1998 May 11.
Article in English | MEDLINE | ID: mdl-9631414

ABSTRACT

Quinoxalinediones such as NBQX are neuroprotective in most models of cerebral ischemia but their poor solubility results in nephrotoxicity limiting their clinical utility. We have investigated the neuroprotective effects of a water soluble AMPA receptor antagonist, YM872, using two in vitro models. The viability of cortical cultures exposed to 400 microM AMPA for 15 min (16.4 +/- 2.6%; n = 10) was significantly (p < 0.05) increased (84.7 +/- 4.6%; n = 6) with YM872 (10 microM) in a concentration-dependent manner. Evoked post-synaptic response amplitudes in oxygen-glucose deprived hippocampal slices treated with 10 microM YM872 (3.5 +/- 0.3 mV; n = 27) were significantly different from untreated deprived slices (0.3 +/- 0.1 mV; n = 31, p < 0.05) and the CA1 neurons appeared viable using a confocal live/dead fluorescence assay with confocal microscopy. The neuroprotection seen with YM872 in vitro warrants further investigation in vivo.


Subject(s)
Hippocampus/physiology , Imidazoles/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Quinoxalines/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/physiology , Excitatory Postsynaptic Potentials/drug effects , Fetus , Glucose/metabolism , Glucose/pharmacology , Hippocampus/drug effects , In Vitro Techniques , Microscopy, Confocal , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
10.
Brain Res Brain Res Protoc ; 2(2): 99-108, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9473610

ABSTRACT

Hippocampal slice models are used to study the mechanisms of ischemia-induced neurotoxicity and to assess the neuroprotective potential of novel therapeutic agents. A number of morphological and functional endpoints are available to assess neuronal viability. The slice model also allows the study of selectively vulnerable neuronal populations within the same preparation. The fluorescence procedure described here provides a method of assessing the viability of neurons in rat hippocampal slices exposed to hypoxic-hypoglycemic conditions. Control and/or treated slices that had been subjected to a 10 min oxygen-glucose deprivation insult are double stained with calcein-AM (4 microM), which stains live cells green, and ethidium homodimer (6 microM), which stains the nucleus of dead cells red. The stained slices are then imaged using confocal microscopy. Vulnerable neurons in the CA1 region of slices deprived of oxygen and glucose became increasingly permeant to ethidium homodimer over the 4 h reperfusion period. Exposure to low Ca2+ concentration (0.3 mM) or the N-, P- and Q-type Ca2+ channel antagonist MVIIC (100 nM), which have been shown to be neuroprotective in this model of ischemia using field evoked post-synaptic potential (EPSP) measures as an endpoint, were also shown to be protective using the fluorescence assay.


Subject(s)
Brain/cytology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Neurons/physiology , Animals , Cell Survival/physiology , Electrophysiology , Glucose/metabolism , Hippocampus/cytology , Hippocampus/physiology , Image Processing, Computer-Assisted , In Vitro Techniques , Male , Oxygen Consumption/physiology , Rats , Rats, Wistar
11.
FASEB J ; 11(13): 1187-97, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9367354

ABSTRACT

The objective of this study was to generate an immortal cell line representative of specialized human brain microvascular endothelia forming the blood-brain barrier (BBB) in vivo. Human capillary and microvascular endothelial cells (HCEC) were transfected with the plasmid pSV3-neo coding for the SV40 large T antigen and the neomycin gene. The neomycin-resistant transfected cells overcame proliferative senescence, and after a 6-8 wk period of crisis produced immortalization-competent cell colonies. Single-cell clones of near-diploid genotype were isolated from these colonies, propagated, and characterized. Immortalized HCEC (SV-HCEC) exhibited accelerated proliferation rates, but remained serum and anchorage dependent and retained the characteristic cobblestone morphology at confluence. SV-HCEC displayed a stable nuclear expression of SV40 large T antigen, lacked the invasiveness of transformed cells, and maintained major phenotypic properties of early passage control cells including expression of factor VIII-related antigen, uptake of acetylated low-density lipoprotein, binding of fluorescently labeled lectins, expression of transferrin receptor and transferrin receptor-mediated endocytosis, and high activities of the BBB-specific enzymes alkaline phosphatase and gamma-glutamyl transpeptidase. The diffusion of radiolabeled sucrose across SV-HCEC monolayers was fivefold lower than that observed with human lung microvascular endothelial cells. Furthermore, media conditioned by fetal human astrocytes increased the transendothelial electrical resistance of SV-HCEC monolayers by 2.5-fold. Therefore, this newly established human cell line expressing the specialized phenotype of BBB endothelium may serve as a readily available in vitro model for studying the properties of the human BBB.


Subject(s)
Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Endothelium, Vascular/physiology , Microcirculation/physiology , Alkaline Phosphatase/metabolism , Animals , Antigens, Viral, Tumor/biosynthesis , Capillaries , Cell Adhesion , Cell Division , Cell Line , Cell Line, Transformed , Cell Transformation, Neoplastic , Clone Cells , Electric Conductivity , Endothelium, Vascular/cytology , Humans , Inulin/pharmacokinetics , Karyotyping , Membrane Potentials , Models, Biological , Rats , Simian virus 40/genetics , Sucrose/pharmacokinetics , Transfection , gamma-Glutamyltransferase/metabolism
12.
Exp Cell Res ; 233(2): 310-20, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9194493

ABSTRACT

The alkaloid protein kinase inhibitor staurosporine induced neuronal cell death with both the morphological and the biochemical characteristics of apoptosis. The punctate chromatin associated with apoptosis with retention of plasma membrane integrity was observed in neurons identified by colocalization of NeuN staining. Such cells had DNA fragmentation visualized by in situ end-labeling which was seen as a laddered pattern upon gel electrophoresis. In contrast cells treated with glutamate did not exhibit either of these morphological or biochemical hallmarks of apoptosis. Instead a much smaller and more compact pyknotic structure was observed associated with smeared DNA fragmentation patterns. A confocal time-lapse study of the appearance of the morphological changes in individual nuclei after staurosporine treatment showed collapse into punctate chromatin over a period of 10 min. In contrast, the collapse into small pyknotic nuclei after glutamate treatment was at least 10 times slower. It is concluded that excitotoxicity produced by glutamate did not induce cell death by an apoptotic mechanism in cultured cortical neurons.


Subject(s)
Apoptosis/drug effects , Cell Death/drug effects , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Glutamates/pharmacology , Neurons/cytology , Staurosporine/pharmacology , Animals , Cells, Cultured , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Protein Kinase C/antagonists & inhibitors , Rats , Time Factors
13.
Brain Res ; 753(2): 209-18, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9125405

ABSTRACT

The presynaptic Ca2+-influx affecting glutamate release during neuropathological processes is mediated via voltage-sensitive calcium channels (VSCCs). There is controversy, however, over the fractional contribution of the specific channel types involved. We have addressed this by investigating the protective effects of various VSCC blockers on oxygen and glucose-deprived rat hippocampal slices. The viability of treated and non-treated slices was assayed electrophysiologically by measuring the evoked population spike (PS) amplitude in the stratum pyramidale of the CA1 region and by imaging slices loaded with fluorochrome dyes specific for dead (ethidium homodimer) and live (calcein) cells using confocal microscopy. PS amplitudes were significantly (P < 0.01) depressed from 4.4 +/- 0.2 mV (n = 38) to 0.2 +/- 0.1 mV (n = 40) after the deprivation insult. Responses from deprived slices treated with omega-conotoxin MVIIC (100 nM; 4.2 +/- 0.5 mV; n = 20) were not significantly different from control, non-deprived slice responses. In contrast, deprived slices treated with either L-type (0.1 or 1 microM nimodipine) or N-type (0.1 or 3 microM omega-conotoxin MVIIA) blockers showed no significant protection. The viability of CA1 neurons as revealed by the fluorescence live/dead confocal viability assay was consistent with the electrophysiological measurements. By comparison with previous studies using P- and Q-type blockers to attempt neuroprotection against the same deprivation insult, the rank order in which specific Ca2+-channel types contribute to neuronal death due to oxygen and glucose deprivation was determined to be Q > N >> P > L.


Subject(s)
Calcium Channels/physiology , Glucose/deficiency , Hippocampus/metabolism , Hippocampus/pathology , Hypoxia/metabolism , Neurons/pathology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/classification , Cell Death , Electrophysiology , Hippocampus/drug effects , Male , Microscopy, Confocal , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar
14.
Brain Res ; 751(2): 206-16, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9099807

ABSTRACT

The objective of this study was to examine the possible role of the cysteine protease cathepsin B (E.C. 3.4.22.1) in the delayed neuronal death in rats subjected to the two-vessel occlusion model of global ischemia. Immunohistochemistry of the hippocampus showed an alteration in the distribution of cathepsin B in CA1 neurons from a lysosomal pattern to a more intense label redistributed into the cytoplasm. This change was not detected until the neurons had become morphologically altered with obvious shrinkage of the cytoplasmic region. Western blotting and enzyme activity measurements of subcellular fractions, including lysosomes and a cell soluble fraction, demonstrated that there was an overall decrease in cathepsin B activity at this time but an increase in the proenzyme form, particularly in the soluble fraction. This was found to be completely different from the marked loss of all forms of cathepsin B in necrotic neurons following decapitation.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cathepsin B/metabolism , Decerebrate State/metabolism , Decerebrate State/physiopathology , Neurons/physiology , Protein Processing, Post-Translational , Animals , Blotting, Western , Brain Ischemia/pathology , Cell Death , Decerebrate State/pathology , Immunohistochemistry , Male , Necrosis , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution
15.
Appl Environ Microbiol ; 63(2): 440-7, 1997 Feb.
Article in English | MEDLINE | ID: mdl-16535509

ABSTRACT

The effects of calcium ions and modulators of calcium movement on Bacillus thuringiensis insecticidal protein toxicity were investigated with Sf9 cells (Spodoptera frugiperda, fall armyworm) by a new B. thuringiensis toxicity assay based on measurement of fluorescence of ethidium homodimer, a high-affinity DNA stain. CryIC toxicity was substantially stimulated by extracellular calcium in a dose-dependent way (in the millimolar range), while toxicity enhancement could not be replicated when calcium was replaced by barium. This incremental toxicity was reduced by cobalt and lanthanum ions, two inorganic-calcium transport inhibitors. Methoxyverapamil, a voltage-dependent calcium channel blocker, and nifedipine, an inhibitor of dihydropyridine-sensitive L-type calcium channels, had no effect on CryIC toxin activity, but BAY K 8644, an L-type calcium channel activator, increased CryIC activity at high concentrations of extracellular calcium. While A23187, a calcium ionophore, and TMB-8, an inhibitor of intracellular-calcium mobilization, did not change CryIC-induced mortality, thapsigargin, an inhibitor of calcium uptake in intracellular stores, and more particularly trifluoperazine, which inhibits calcium-calmodulin-dependent processes, increased CryIC-mediated toxicity. The incremental effect of extracellular calcium on CryIC-induced toxicity was consistent with an increased concentration of intracellular calcium.

16.
Neuropharmacology ; 36(1): 107-13, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9144647

ABSTRACT

We investigated the neuroprotective efficacy of the P-type Ca2+ channel antagonist daurisoline against electroshock-induced convulsions in rats and mice, hypoxic/hypoglycemic-induced damage in rat hippocampal slices and brain damage induced by occlusion of the middle cerebral artery (MCA) in rats. Daurisoline applied intravenously (i.v.) (bolus of 1-60 mg/kg) reduced the spontaneous activity of rat cerebellar Purkinje cells in a dose-dependent manner, a result demonstrating activity in the brain with systemic administration of the compound. While this effect reversed rapidly in about 10-20 min following bolus-application of the drug at doses of up to 30 mg/kg, a dose of 60 mg/kg consistently induced a depression of respiration followed by death of the animals. Daurisoline administered at 10-30 mg/kg did not prevent electroshock-induced convulsions in mice or rats, nor did it reduce the neuronal damage in hippocampal slices induced by a hypoxic/hypoglycemic insult in vitro by MCA occlusion in vivo. These observations do not support the hypothesis that P-type Ca2+ channels are promising drug targets for the acute treatment of epileptic convulsions and/or ischemic stroke.


Subject(s)
Alkaloids/therapeutic use , Anticonvulsants/therapeutic use , Benzylisoquinolines , Brain Ischemia/drug therapy , Calcium Channel Blockers/therapeutic use , Epilepsy/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/pathology , Evoked Potentials/drug effects , Male , Mice , Microscopy, Confocal , Purkinje Cells/drug effects , Rats
17.
Neuropharmacology ; 35(8): 1037-48, 1996.
Article in English | MEDLINE | ID: mdl-9121606

ABSTRACT

The efficacy and mechanisms of 1-amino-cyclopentyl-1S,3R-dicarboxylate (1S,3R-ACPD)-induced neuroprotection were investigated in rat hippocampal slices subjected to 10 min of oxygen and glucose deprivation. Neuronal viability was assessed by measuring both the amplitude of evoked population spike in the CA1 pyramidale and by imaging CA1 neurons using a live/dead fluorescence assay with confocal microscopy. CA1 pyramidal neurons in oxygen-glucose deprived slices remained viable for up to 120 min following the insult but were dead by 240 min. Pretreatment with 1S,3R-ACPD significantly protected the oxygen-glucose deprived slices in a concentration-dependent fashion. Oxygen-glucose deprived slices pretreated for the same period with the protein kinase C (PKC) activation phorbol 12-myristate 13-acetate (PMA; 1 microM) were significantly protected whereas oxygen-glucose deprived slices treated with the adenylyl cyclase activator, forskolin (30 microM) were not. Oxygen-glucose deprivation induced a rapid and persistent decrease (approximately 50%) in PKC activity and a > 6 fold increase in cyclic adenosine monophosphate (cAMP) levels in whole hippocampal slices. While 1S,3R-ACPD did not stimulate PKC activity and had no effect on basal cAMP in whole slices, it significantly enhanced the rate of return of cAMP to basal levels following reperfusion. Consistent with this observation, the 1S,3R-ACPD-induced neuroprotection was inhibited by forskolin (30 microM). These results suggest that in vitro neuroprotection of CA1 neurons by 1S,3R-ACPD involves metabotropic glutamate receptors negatively linked to cAMP and possibly those which increase PKC activity.


Subject(s)
Cycloleucine/analogs & derivatives , Glucose/physiology , Hippocampus/drug effects , Hypoxia, Brain/pathology , Neuroprotective Agents/pharmacology , Animals , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Cycloleucine/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Hippocampus/enzymology , Hippocampus/pathology , Hypoxia, Brain/enzymology , In Vitro Techniques , Male , Microscopy, Confocal , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Signal Transduction/drug effects
18.
Neuroreport ; 6(12): 1617-20, 1995 Aug 21.
Article in English | MEDLINE | ID: mdl-8527726

ABSTRACT

Excessive accumulation of Ca2+ in neurones and glutamate release are involved in neuropathological processes, including ischaemia. We investigated the neuroprotective effects of the Ca2+ channel antagonist, omega-Aga-IVA, in CA1 pyramidal neurones in rat hippocampal slices following an in vitro hypoxic-hypoglycaemic insult. Following this insult, evoked post-synaptic response amplitudes decreased from 3.7 +/- 0.5 mV to 0.6 +/- 0.2 mV and the CA1 neurones appeared dead using a live/dead fluorescence assay with confocal microscopy. Slices treated with 200 nM omega-Aga-IVA had evoked response amplitudes not significantly different from control (3.3 +/- 0.5 mV) and the CA1 neurones appeared viable using the live/dead fluorescence assay. The neuroprotective efficacy of omega-Aga-IVA suggests that omega-Aga-IVA-sensitive Ca2+ channels participate in ischaemic neuronal death and constitute a potential target of therapeutic intervention.


Subject(s)
Brain Ischemia/drug therapy , Calcium Channel Blockers/pharmacology , Hippocampus/drug effects , Spider Venoms/pharmacology , Animals , Electrophysiology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Microscopy, Electron , Rats , Rats, Sprague-Dawley , omega-Agatoxin IVA
20.
Pflugers Arch ; 427(5-6): 500-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7971148

ABSTRACT

A linker insertion mutagenesis strategy was developed to probe functional subdomains of the RIIA Na+ channel alpha-subunit. We describe mutations within the first two repeat domains that provide new functional information for three segments of the channel structure. 1. The insertion of two alanine residues within the short peptide segment joining helices S4 and S5 in domain II had two effects: a depolarizing shift of steady-state activation and reduced single-channel currents. These results suggest that the peptide segment following the S4 voltage sensor is involved in the activation process and is facing the ion pore. 2. An insertion immediately N-terminal to the proposed transmembrane helix S1 in domain II shifted the steady-state activation in the depolarizing direction, suggesting a functional role in channel gating. 3. Insertions in the large, cytoplasmic loop between domains I and II affect two channel functions: inactivation and protein kinase A modulation. These results demonstrate that the linker insertion approach can provide novel insights into the structure-function relationships of large, multi-domain ion channel proteins.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Ion Channel Gating/physiology , Mutagenesis, Insertional , Sodium Channels/genetics , Sodium Channels/metabolism , Amino Acid Sequence , Animals , Base Sequence , Molecular Sequence Data , Patch-Clamp Techniques , Protein Kinase C/genetics , Protein Kinase C/metabolism , Repetitive Sequences, Nucleic Acid , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL