Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Neuroinflammation ; 18(1): 168, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34332596

ABSTRACT

BACKGROUND: Following stroke, changes in neuronal connectivity in tissue surrounding the infarct play an important role in both spontaneous recovery of neurological function and in treatment-induced improvements in function. Microglia and astrocytes influence this process through direct interactions with the neurons and as major determinants of the local tissue environment. Subpopulations of peri-infarct glia proliferate early after stroke providing a possible target to modify recovery. Treatment with cell cycle inhibitors can reduce infarct volume and improve functional recovery. However, it is not known whether these inhibitors can influence neurological function or alter the responses of peri-infarct glia without reducing infarction. The present study aimed to address these issues by testing the effects of the cell cycle inhibitor, olomoucine, on recovery and peri-infarct changes following photothrombotic stroke. METHODS: Stroke was induced by photothrombosis in the forelimb sensorimotor cortex in Sprague-Dawley rats. Olomoucine was administered at 1 h and 24 h after stroke induction. Forelimb function was monitored up to 29 days. The effects of olomoucine on glial cell responses in peri-infarct tissue were evaluated using immunohistochemistry and Western blotting. RESULTS: Olomoucine treatment did not significantly affect maximal infarct volume. Recovery of the affected forelimb on a placing test was impaired in olomoucine-treated rats, whereas recovery in a skilled reaching test was substantially improved. Olomoucine treatment produced small changes in aspects of Iba1 immunolabelling and in the number of CD68-positive cells in cerebral cortex but did not selectively modify responses in peri-infarct tissue. The content of the astrocytic protein, vimentin, was reduced by 30% in the region of the lesion in olomoucine-treated rats. CONCLUSIONS: Olomoucine treatment modified functional recovery in the absence of significant changes in infarct volume. The effects on recovery were markedly test dependent, adding to evidence that skilled tasks requiring specific training and general measures of motor function can be differentially modified by some interventions. The altered recovery was not associated with specific changes in key responses of peri-infarct microglia, even though these cells were considered a likely target for early olomoucine treatment. Changes detected in peri-infarct reactive astrogliosis could contribute to the altered patterns of functional recovery.


Subject(s)
Astrocytes/drug effects , Kinetin/pharmacology , Microglia/drug effects , Motor Cortex/drug effects , Recovery of Function/drug effects , Stroke/physiopathology , Animals , Cell Cycle/drug effects , Disease Models, Animal , Gliosis/pathology , Gliosis/physiopathology , Male , Microglia/pathology , Motor Cortex/pathology , Motor Cortex/physiopathology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Stroke/pathology
2.
J Neuroinflammation ; 16(1): 6, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30626393

ABSTRACT

BACKGROUND: Altered neuronal connectivity in peri-infarct tissue is an important contributor to both the spontaneous recovery of neurological function that commonly develops after stroke and improvements in recovery that have been induced by experimental treatments in animal models. Microglia and astrocytes are primary determinants of the environment in peri-infarct tissue and hence strongly influence the potential for neuronal plasticity. However, the specific roles of these cells and the timing of critical changes in their function are not well understood. Minocycline can protect against ischemic damage and promote recovery. These effects are usually attributed, at least partially, to the ability of this drug to suppress microglial activation. This study tested the ability of minocycline treatment early after stroke to modify reactive responses in microglia and astrocytes and improve recovery. METHODS: Stroke was induced by photothrombosis in the forelimb sensorimotor cortex of Sprague-Dawley rats. Minocycline was administered for 2 days after stroke induction and the effects on forelimb function assessed up to 28 days. The responses of peri-infarct Iba1-positive cells and astrocytes were evaluated using immunohistochemistry and Western blots. RESULTS: Initial characterization showed that the numbers of Iba1-positive microglia and macrophages decreased in peri-infarct tissue at 24 h then increased markedly over the next few days. Morphological changes characteristic of activation were readily apparent by 3 h and increased by 24 h. Minocycline treatment improved the rate of recovery of motor function as measured by a forelimb placing test but did not alter infarct volume. At 3 days, there were only minor effects on core features of peri-infarct microglial reactivity including the morphological changes and increased density of Iba1-positive cells. The treatment caused a decrease of 57% in the small subpopulation of cells that expressed CD68, a marker of phagocytosis. At 7 days, the expression of glial fibrillary acidic protein and vimentin was markedly increased by minocycline treatment, indicating enhanced reactive astrogliosis. CONCLUSIONS: Early post-stroke treatment with minocycline improved recovery but had little effect on key features of microglial activation. Both the decrease in CD68-positive cells and the increased activation of astrogliosis could influence neuronal plasticity and contribute to the improved recovery.


Subject(s)
Astrocytes/drug effects , Brain Infarction , Microglia/drug effects , Minocycline/therapeutic use , Recovery of Function/drug effects , Stroke/complications , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain Infarction/drug therapy , Brain Infarction/etiology , Brain Infarction/pathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Forelimb/physiopathology , Intracranial Thrombosis/complications , Male , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Stroke/drug therapy , Stroke/etiology , Stroke/pathology , Time Factors
3.
Neurochem Res ; 43(3): 637-649, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29330684

ABSTRACT

Treatment with mature brain-derived neurotrophic factor (mBDNF) promotes functional recovery after ischemia in animal trials but the possible role of its precursor protein proBDNF and its receptors or the factors responsible for the conversion of proBDNF to mBDNF in ischemic stroke are not known. The main aim of this study was to characterize the time-dependent expression of genes and/or proteins related to BDNF processing and signaling after ischemia as well as the sensorimotor behavioral dysfunction in a photothrombotic ischemic model in rats. Characterization of different genes and proteins related to BDNF processing and signaling was performed using qPCR, immunoblotting and enzyme-linked immunosorbent assays. We showed in this study that some sensory and motor functional deficiencies appeared in the ischemic group at day 1 and persisted until day 14. Most changes in gene expression of BDNF and its processing enzymes occurred within the first 24 h in the ipsilateral cortex, but not in the contralateral cortex. At the protein level, proBDNF expression was increased at 6 h, mBDNF expression was increased between 15 h and 1 day while p75 receptor protein expression was increased between 6 h and 3 days in the ipsilateral cortex, but not in the contralateral cortex. Therefore, cerebral ischemia in rats led to the up-regulation of genes and/or proteins of BDNF, proBDNF and their processing enzymes and receptors in a time-dependent manner. We propose that the balance between BDNF and proBDNF and their associated proteins may play an important role in the pathogenesis and recovery from ischemia.


Subject(s)
Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Animals, Newborn , Protein Precursors/metabolism , Rats, Sprague-Dawley , Signal Transduction/physiology , Up-Regulation
4.
Biochim Biophys Acta ; 1802(1): 80-91, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19751827

ABSTRACT

Stroke most commonly results from occlusion of a major artery in the brain and typically leads to the death of all cells within the affected tissue. Mitochondria are centrally involved in the development of this tissue injury due to modifications of their major role in supplying ATP and to changes in their properties that can contribute to the development of apoptotic and necrotic cell death. In animal models of stroke, the limited availability of glucose and oxygen directly impairs oxidative metabolism in severely ischemic regions of the affected tissue and leads to rapid changes in ATP and other energy-related metabolites. In the less-severely ischemic "penumbral" tissue, more moderate alterations develop in these metabolites, associated with near normal glucose use but impaired oxidative metabolism. This tissue remains potentially salvageable for at least the first few hours following stroke onset. Early restoration of blood flow can result in substantial recovery of energy-related metabolites throughout the affected tissue. However, glucose oxidation is markedly decreased due both to lower energy requirements in the post-ischemic tissue and limitations on the mitochondrial oxidation of pyruvate. A secondary deterioration of mitochondrial function subsequently develops that may contribute to progression to cell loss. Mitochondrial release of multiple apoptogenic proteins has been identified in ischemic and post-ischemic brain, mostly in neurons. Pharmacological interventions and genetic modifications in rodent models strongly implicate caspase-dependent and caspase-independent apoptosis and the mitochondrial permeability transition as important contributors to tissue damage, particularly when induced by short periods of temporary focal ischemia.


Subject(s)
Mitochondria/metabolism , Stroke/metabolism , Animals , Brain Ischemia/metabolism , Cell Death/physiology , Energy Metabolism , Humans , Models, Biological , Oxygen/metabolism , Reperfusion Injury
5.
J Neurochem ; 119(6): 1253-61, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21985402

ABSTRACT

Mitochondria are key contributors to many forms of cell death including those resulting from neonatal hypoxic-ischemic brain injury. Mice have become increasingly popular in studies of brain injury, but there are few reports evaluating mitochondrial isolation procedures for the neonatal mouse brain. Using evaluation of respiratory activity, marker enzymes, western blotting and electron microscopy, we have compared a previously published procedure for isolating mitochondria from neonatal mouse brain (method A) with procedures adapted from those for adult rats (method B) and neonatal rats (method C). All three procedures use Percoll density gradient centrifugation as a key step in the isolation but differ in many aspects of the fractionation procedure and the solutions used during fractionation. Methods A and B both produced highly enriched fractions of well-coupled mitochondria with high rates of respiratory activity. The fraction from method C exhibited less preservation of respiratory properties and was more contaminated with other subcellular components. Method A offers the advantage of being more rapid and producing larger mitochondrial yields making it useful for routine applications. However, method B produced mitochondria that were less contaminated with synaptosomes and associated cytosolic components that suits studies that have a requirement for higher mitochondrial purification.


Subject(s)
Brain/ultrastructure , Mitochondria/ultrastructure , Adenosine Diphosphate/pharmacology , Animals , Animals, Newborn , Electron Transport Complex IV/metabolism , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/metabolism , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/ultrastructure
6.
Cell Transplant ; 30: 963689720984437, 2021.
Article in English | MEDLINE | ID: mdl-33432826

ABSTRACT

Dental pulp contains multipotent mesenchymal stem cells that improve outcomes when administered early after temporary middle cerebral artery occlusion in rats. To further assess the therapeutic potential of these cells, we tested whether functional recovery following stroke induced by photothrombosis could be modified by a delayed treatment that was initiated after the infarct attained maximal volume. Photothrombosis induces permanent focal ischemia resulting in tissue changes that better reflect key aspects of the many human strokes in which early restoration of blood flow does not occur. Human dental pulp stem cells (approximately 400 × 103 viable cells) or vehicle were injected into the infarct and adjacent brain tissue of Sprague-Dawley rats at 3 days after the induction of unilateral photothrombotic stroke in the sensorimotor cortex. Forepaw function was tested up to 28 days after stroke. Cellular changes in peri-infarct tissue at 28 days were assessed using immunohistochemistry. Rats treated with the stem cells showed faster recovery compared with vehicle-treated animals in a test of forelimb placing in response to vibrissae stimulation and in first attempt success in a skilled forelimb reaching test. Total success in the skilled reaching test and forepaw use during exploration in a Perspex cylinder were not significantly different between the 2 groups. At 28 days after stroke, rats treated with the stem cells showed decreased immunolabeling for glial fibrillary acidic protein in tissue up to 1 mm from the infarct, suggesting decreased reactive astrogliosis. Synaptophysin, a marker of synapses, and collagen IV, a marker of capillaries, were not significantly altered at this time by the stem-cell treatment. These results indicate that dental pulp stem cells can accelerate recovery without modifying initial infarct formation. Decreases in reactive astrogliosis in peri-infarct tissue could have contributed to the change by promoting adaptive responses in neighboring neurons.


Subject(s)
Astrocytes/metabolism , Dental Pulp/metabolism , Recovery of Function/physiology , Stem Cell Transplantation/methods , Stem Cells/metabolism , Stroke/physiopathology , Stroke/therapy , Animals , Disease Models, Animal , Humans , Male , Rats , Rats, Sprague-Dawley
7.
J Neurochem ; 109 Suppl 1: 101-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19393015

ABSTRACT

Glutathione in the mitochondria is an important determinant of cellular responses to oxidative stress. Mitochondrial glutathione is maintained by uptake from the cytosol, a process that has been little studied in brain cells. In the present study, measurements using isolated rat brain mitochondria showed a rapid uptake of [3H]-glutathione that was strongly influenced by the mitochondrial glutathione content. [3H]-glutathione incorporated into the mitochondria was not rapidly released. Uptake was inhibited by substrates and inhibitors for several known mitochondrial anion transporters. Citrate, isocitrate and benzene-1,2,3-tricarboxylate were particularly effective inhibitors, suggesting a possible role for a tricarboxylate carrier in the glutathione transport. The properties of uptake differed greatly from those reported previously for mitochondria from kidney and liver. In astrocytes in primary culture, diethylmaleate or hydrogen peroxide treatment resulted in depletion of cytosolic and mitochondrial glutathione. The pattern of restoration of glutathione content in the presence of glutathione precursors following treatment with diethylmaleate was consistent with uptake into mitochondria being controlled primarily by the glutathione gradient between the cytosol and mitochondria. However, following hydrogen peroxide treatment, recovery of glutathione in the mitochondria initially preceded comparable proportional restoration in the cytosol, suggesting the possibility of additional controls on glutathione uptake in some conditions.


Subject(s)
Astrocytes/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Animals , Brain/cytology , Brain Chemistry/drug effects , Brain Chemistry/physiology , Cells, Cultured , Hydrogen Peroxide/metabolism , Maleates/pharmacology , Mitochondrial Membranes/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
8.
Neurochem Res ; 34(10): 1857-66, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19488856

ABSTRACT

Mitochondria isolated from brain tissue following middle cerebral artery occlusion or during early reperfusion were tested for their ability to generate a membrane potential under standard conditions in vitro. Membrane potential was evaluated based on rhodamine 123 fluorescence in the mitochondria as detected using flow cytometry. Compared with equivalent samples from the contralateral hemisphere, the geometric mean fluorescence was significantly lower in mitochondria prepared from the striatum and perifocal tissue in the cortex at 3 h ischemia. During reperfusion, this property was decreased in mitochondria from tissue in the striatum and cortex that had been part of severely ischemic core tissue during the arterial occlusion. These findings provide additional evidence that mitochondria develop changes during ischemia and reperfusion that are likely to limit their ability to respond to changing energy requirements and contribute to cell dysfunction and cell death. It also demonstrates the ability to gain a sensitive measure of these mitochondrial changes using flow cytometry.


Subject(s)
Brain Ischemia/physiopathology , Brain/physiology , Cell Separation/methods , Flow Cytometry/methods , Intracellular Membranes/pathology , Mitochondria/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Animals , Brain/blood supply , Brain/pathology , Brain Ischemia/pathology , Intracellular Membranes/physiology , Male , Membrane Potentials/physiology , Rats , Rats, Sprague-Dawley
9.
Neurochem Int ; 107: 88-103, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28057555

ABSTRACT

Alterations in neuronal connectivity, particularly in the "peri-infarct" tissue adjacent to the region of ischemic damage, are important contributors to the spontaneous recovery of function that commonly follows stroke. Peri-infarct astrocytes undergo reactive astrogliosis and play key roles in modulating the adaptive responses in neurons. This reactive astrogliosis shares many features with that induced by other forms of damage to the central nervous system but also differs in details that potentially influence neurological recovery. A subpopulation of astrocytes within a few hundred micrometers of the infarct proliferate and are centrally involved in the development of the glial scar that separates the damaged tissue in the infarct from surrounding normal brain. The intertwined processes of astrocytes adjacent to the infarct provide the core structural component of the mature scar. Interventions that cause early disruption of glial scar formation typically impede restoration of neurological function. Marked reactive astrogliosis also develops in cells more distant from the infarct but these cells largely remain in the spatial territories they occupied prior to stroke. These cells play important roles in controlling the extracellular environment and release proteins and other molecules that are able to promote neuronal plasticity and improve functional recovery. Treatments manipulating aspects of reactive astrogliosis can enhance neuronal plasticity following stroke. Optimising these treatments for use in human stroke would benefit from a more complete characterization of the specific responses of peri-infarct astrocytes to stroke as well as a better understanding of the influence of other factors including age, sex, comorbidities and reperfusion of the ischemic tissue.


Subject(s)
Astrocytes/metabolism , Gliosis/metabolism , Recovery of Function/physiology , Stroke/metabolism , Animals , Astrocytes/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Gliosis/pathology , Humans , Stroke/pathology
10.
J Neurosci ; 24(37): 8019-28, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15371502

ABSTRACT

Glutathione, a major endogenous antioxidant, is found in two intracellular pools in the cytoplasm and the mitochondria. To investigate the importance of the smaller mitochondrial pool, we developed conditions based on treatment with ethacrynic acid that produced near-complete and highly selective depletion of mitochondrial glutathione in cultured astrocytes. Recovery of mitochondrial glutathione was only partial over several hours, suggesting slow net uptake from the cytoplasm. Glutathione depletion alone did not significantly affect mitochondrial membrane potential, ATP content, or cell viability when assessed after 24 hr, although the activities of respiratory chain complexes were altered. However, these astrocytes showed a greatly enhanced sensitivity to 3-morpholinosydnonimine, a peroxynitrite generator. Treatment with 200 microm 3-morpholinosydnonimine produced decreases within 3 hr in mitochondrial membrane potential and ATP content and caused the release of lactate dehydrogenase, contrasting with preservation of these properties in control cells. These properties deteriorated further by 24 hr in the glutathione-depleted cells and were associated with morphological changes indicative of necrotic cell death. This treatment enhanced the alterations in activities of the respiratory chain complexes observed with glutathione depletion alone. Cell viability was markedly improved by cyclosporin A, suggesting a role for the mitochondrial permeability transition in the astrocytic death. These studies provide the most direct evidence available for any cell type on the roles of mitochondrial glutathione. They demonstrate the critical importance of this metabolite pool in protecting against peroxynitrite-induced damage in astrocytes and indicate a key contribution in determining the activities of respiratory chain components.


Subject(s)
Astrocytes/drug effects , Glutathione/deficiency , Mitochondria/drug effects , Molsidomine/analogs & derivatives , Peroxynitrous Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Astrocytes/physiology , Brain/cytology , Cell Death/drug effects , Cells, Cultured/drug effects , Cells, Cultured/physiology , Citrate (si)-Synthase/analysis , Cyclosporine/pharmacology , Cytoplasm/metabolism , Electron Transport/drug effects , Electron Transport Complex I/physiology , Ethacrynic Acid/pharmacology , Glutathione/physiology , Intracellular Membranes/physiology , L-Lactate Dehydrogenase/analysis , Membrane Potentials/drug effects , Mitochondria/physiology , Molsidomine/pharmacology , Molsidomine/toxicity , Oxidative Stress , Rats , Rats, Sprague-Dawley
11.
J Cereb Blood Flow Metab ; 25(4): 440-50, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15674239

ABSTRACT

Astrocytes play many roles essential for normal brain activity. The ability of these cells to recover after temporary focal cerebral ischemia is likely to be one important determinant of the extent of brain dysfunction and tissue damage. We have assessed astrocytic function based on the incorporation of radiolabel from 1-14C-acetate into glutamine at 1 hour of recirculation after middle cerebral artery occlusion for 2 or 3 hours in rats. There were marked differences in the response between subregions within the tissue subjected to ischemia, but the overall pattern of changes was similar after each ischemic period. The striatum, which forms part of the severely ischemic focal tissue during arterial occlusion, showed a large (44% to 68%) decrease in glutamine labeling compared with equivalent tissue from the contralateral hemisphere. In contrast, 14C-glutamine content was not significantly altered in perifocal tissue in the cerebral cortex, which was subjected to more moderate ischemia. Cortical focal tissue also was not significantly affected, but the response was much more variable between rats. In these brain subregions, the extent of recovery of the 14C-acetate metabolism after ischemia was not a good predictor of the likelihood of subsequent infarct development. Interestingly, a similar pattern of responses persisted when recirculation was extended to 4 hours. These results indicate that many astrocytes, particularly in the cortex, remain viable and capable of at least some complex oxidative metabolism during the first few hours of recirculation.


Subject(s)
Acetates/metabolism , Astrocytes/metabolism , Ischemic Attack, Transient/metabolism , Animals , Astrocytes/drug effects , Blood Gas Analysis , Cerebral Cortex/pathology , Glutamic Acid/metabolism , Glutamine/metabolism , Hemodynamics/physiology , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/physiopathology , Male , Neostriatum/pathology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
12.
Neurosci Lett ; 382(3): 227-30, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15925095

ABSTRACT

This study examined the neuroprotective effects and possible hepatotoxicity of (-)-epigallocatechin gallate (EGCG) in a rat model of transient focal cerebral ischemia. Male Sprague-Dawley rats (265-295 g) were treated with either 50 mg kg(-1) of EGCG or saline, i.p., immediately post-ischemia and every day thereafter, in a middle cerebral artery occlusion model of stroke. Sacrifice occurred 72 h post-ischemia and 2,3,5-triphenyltetrazolium chloride staining was used to quantify neuronal infarction. Hepatotoxicity was determined by taking blood samples for plasma alanine aminotransferase (ALT) activity. Spleen, kidney, liver and testes wet weights were also recorded. Total infarct volume was significantly (P<0.05) reduced in the EGCG-treated group as compared to controls. Analysis of the mean infarct area showed a significant (P<0.05) decrease in slices 6 and 7 in the EGCG-treated group. No significant differences were found in organ weights or ALT levels between treatment groups. Our findings, in part, validate and extend previous observations illustrating that 50 mg kg(-1), i.p. EGCG is non-toxic and neuroprotective. However, we also found that EGCG treatment appreciably increased (>50%) the number of animals that developed an intracerebral hemorrhage. We therefore conclude that 50 mg kg(-1) EGCG is not a viable intervention for the acute treatment of cerebral ischemia, as it is likely to increase the risk of intracerebral hemorrhaging.


Subject(s)
Brain Ischemia/drug therapy , Brain/pathology , Catechin/analogs & derivatives , Catechin/therapeutic use , Neuroprotective Agents/therapeutic use , Alanine Transaminase/drug effects , Alanine Transaminase/metabolism , Animals , Brain/drug effects , Brain Ischemia/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Liver/drug effects , Male , Rats , Rats, Sprague-Dawley
13.
Neurochem Int ; 40(6): 511-26, 2002 May.
Article in English | MEDLINE | ID: mdl-11850108

ABSTRACT

Tissue infarction, involving death of essentially all cells within a part of the brain, is a common pathology resulting from stroke and an important determinant of the long-term consequences of this disorder. The cell death that leads to infarct formation is likely to be the result of multiple interacting pathological processes. A range of factors, including the severity of the ischemic insult and whether this is permanent or reversed, determine which mechanisms predominate. Although evaluating mitochondrial properties in intact brain is difficult, evidence for several potentially deleterious responses to cerebral ischemia or post-ischemic reperfusion have been obtained from investigations using animal models of stroke. Marked changes in ATP and related energy metabolites develop quickly in response to occlusion of a cerebral artery, as expected from limitations in the delivery of oxygen and glucose. However, these alterations are often only partially reversed on reperfusion despite improved substrate delivery. Ischemia-induced decreases in the mitochondrial capacity for respiratory activity probably contribute to the ongoing impairment of energy metabolism during reperfusion and possibly also to the magnitude of changes seen during ischemia. Conditions during reperfusion are likely to be conducive to the induction of the permeability transition in mitochondria. There are as yet no well-characterized techniques to identify this change in the intact brain. However, the protective effects of some agents that block formation of the transition pore are consistent with both the induction of the permeability transition during early recirculation and a role for this in the development of tissue damage. Release of cytochrome c into the cytoplasm of cells has been observed with both permanent and reversed ischemia and could trigger the death of some cells by apoptosis, a process which probably contributes to the expansion of the ischemic lesion. Mitochondria are also likely to contribute to the widely-accepted role of nitric oxide in the development of ischemic damage. These organelles are a probable target for the deleterious effects of this substance and can also act as a source of superoxide for reaction with the nitric oxide to produce the damaging species, peroxynitrite. Further characterization of these mitochondrial responses should help to elucidate the mechanisms of cell death due to cerebral ischemia and possibly point to novel sites for therapeutic interventions in stroke.


Subject(s)
Mitochondria/pathology , Stroke/pathology , Animals , Brain/pathology , Brain Ischemia/pathology , Cell Death/physiology , Energy Metabolism/physiology , Free Radicals/metabolism , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Oxygen Consumption/physiology , Reperfusion Injury/pathology
14.
Neurochem Int ; 44(3): 153-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14568558

ABSTRACT

Glutathione is a central component in the antioxidant defences of cells. We have recently reported an early and selective loss of total (reduced plus oxidised) glutathione from mitochondria isolated from rat brain following occlusion of the middle cerebral artery. This mitochondrial glutathione depletion showed an apparent association with the tissue damage that developed during subsequent reperfusion, suggesting that it could be an important determinant of susceptibility to cell loss. In the present study, we have investigated whether in vivo treatment with glutathione ethyl ester can modulate mitochondrial glutathione in the brain and whether this treatment can influence the response to focal ischemia. In further support of our previous findings, middle cerebral artery occlusion caused a duration-dependent partial loss of mitochondrial glutathione. Bilateral injections of glutathione ethyl ester immediately prior to induction of unilateral focal ischemia resulted in a substantial increase in glutathione in mitochondria from the striatum of both the non-ischemic hemisphere (190% of saline-treated controls) and the ischemic hemisphere (240% of controls) at 2h after arterial occlusion. Total tissue glutathione was not affected by the ester treatment at this time. A smaller increase in mitochondrial glutathione was observed at 3h of occlusion in the non-ischemic striatum following ester treatment but at this time point glutathione was not significantly altered in mitochondria from the ischemic hemisphere. Pre-ischemic treatment with glutathione ester did not significantly change the volume of tissue infarction assessed at 48 h following ischemia for 2 or 3h. These studies demonstrate that glutathione ethyl ester is a highly effective modulator of the mitochondrial glutathione pool in the intact brain and provides a useful means for further investigating the role of this antioxidant in the development of tissue damage in ischemia and other brain disorders.


Subject(s)
Brain Ischemia/metabolism , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione/pharmacology , Mitochondria/metabolism , Animals , Brain Chemistry/drug effects , Brain Chemistry/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Functional Laterality/physiology , Male , Middle Cerebral Artery/physiology , Mitochondria/drug effects , Neostriatum/pathology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
15.
Brain Res ; 989(2): 221-30, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-14556944

ABSTRACT

The ability of glia to recover essential functions following a period of focal cerebral ischemia is likely to be one important factor influencing the severity of tissue damage that subsequently develops. In this study, we have compared changes in immunoreactivity of markers specific for astrocytes, NG2-positive glia and neurons in tissue subregions during early reperfusion following 3 h of middle cerebral artery occlusion to provide insights into possible differential susceptibility of these cell populations. Under the conditions used, infarction ultimately encompasses most of the perfusion territory of the occluded artery. Nonetheless, alterations in immunoreactivity during the first 3 h of recirculation were restricted to brain regions that had been subjected to severe ischemia. In the striatum, cellular immunoreactivity for NG2 and neuronal markers, NeuN and microtubule-associated protein 2, was greatly reduced by 1 h of reperfusion and declined further at 3 h. NG2 labeling of blood vessels in the striatum appeared post-ischemically, mimicking expression of this protein during development. Less severe changes were seen in the neuronal markers in overlying cerebral cortex. In contrast to the losses of other cellular proteins, immunoreactivity for the astrocytic marker, glial fibrillary acidic protein, was preserved in all tissue that had been subjected to severe ischemia and labeling of another astrocytic protein, glutamine synthetase, was increased by 3 h of reperfusion. These findings provide the first evidence of marked sensitivity of NG2-immunoreactivity to severe ischemia and suggest a greater initial resistance of astrocytes compared with neurons and NG2-positive glia to ischemia-reperfusion damage.


Subject(s)
Antigens/metabolism , Astrocytes/metabolism , Brain Ischemia/metabolism , Phosphopyruvate Hydratase/metabolism , Proteoglycans/metabolism , Reperfusion Injury/metabolism , Animals , Benzimidazoles/metabolism , Biomarkers , Blood Glucose , Blood Pressure , Brain/cytology , Brain/metabolism , Energy Metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Quinolinium Compounds/metabolism , Rats , Rats, Sprague-Dawley , Thiazoles/metabolism
16.
Neurosci Lett ; 354(2): 163-5, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14698463

ABSTRACT

Oxidative stress plays an important role in the development of tissue damage following transient focal cerebral ischaemia. Glutathione is a central component in the antioxidant defence of cells. We have previously shown a close association between mitochondrial glutathione loss and cell death following middle cerebral artery (MCA) occlusion. Glutathione monoethyl ester increases cellular glutathione and is particularly effective in increasing the mitochondrial pool. In the present investigation, we infused glutathione monoethyl ester into the third ventricle during 2 h of MCA occlusion and 48 h of reperfusion. Infarct size was reduced from 46% of the total ischaemic hemisphere in saline-treated animals to 16% following ester treatment. Thus, glutathione monoethyl ester provides neuroprotection following transient focal cerebral ischaemia.


Subject(s)
Cerebral Infarction/prevention & control , Glutathione/analogs & derivatives , Glutathione/pharmacology , Ischemic Attack, Transient/drug therapy , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Glutathione/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/physiopathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/physiopathology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
17.
Mitochondrion ; 12(4): 465-71, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22735573

ABSTRACT

We have previously reported a heteroplasmic mtDNA mutation (T1095C) in the 12SrRNA gene of an Italian family with features of maternally-inherited parkinsonism, antibiotic-mediated deafness and peripheral neuropathy. In the present study, we demonstrate that a transmitochondrial cybrid line derived from the proband of this family shows selective depletion of mitochondrial glutathione and decreases in the activity of complex II/III. Moreover, when exposed to an aminoglycoside antibiotic these cells responded with a ten-fold increase in the number of apoptotic cells compared to controls. These results support a pathogenic role for the T1095C mutation and indicate that the mutation increases the risk for aminoglycoside-induced toxicity.


Subject(s)
Anti-Bacterial Agents/adverse effects , Apoptosis , DNA, Mitochondrial/genetics , Gentamicins/adverse effects , Mitochondria/drug effects , Mitochondria/genetics , Point Mutation , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Cell Line , Genes, rRNA , Gentamicins/metabolism , Gentamicins/toxicity , Humans , Italy , Mitochondria/metabolism , RNA, Ribosomal/genetics
18.
Nat Protoc ; 3(7): 1228-39, 2008.
Article in English | MEDLINE | ID: mdl-18600228

ABSTRACT

We have developed procedures that combine differential centrifugation and discontinuous Percoll density gradient centrifugation to isolate mitochondria from rat forebrains and brain subregions. The use of Percoll density gradient centrifugation is central to obtaining preparations that contain little contamination with synaptosomes and myelin. Protocols are presented for three variations of this procedure that differ in their suitability for dealing with large or small samples, in the proportion of total mitochondria isolated and in the total preparation time. One variation uses digitonin to disrupt synaptosomes before mitochondrial isolation. This method is well suited for preparing mitochondria from small tissue samples, but the isolated organelles are not appropriate for all studies. Each of the procedures produces mitochondria that are well coupled and exhibit high rates of respiratory activity. The procedures require an initial setup time of 45-75 min and between 1 and 3 h for the mitochondrial isolation.


Subject(s)
Brain/cytology , Cell Fractionation/methods , Centrifugation, Density Gradient/methods , Mitochondria , Animals , Povidone , Rats , Silicon Dioxide
19.
Neurochem Res ; 32(4-5): 663-70, 2007.
Article in English | MEDLINE | ID: mdl-17024570

ABSTRACT

Nitric oxide has been strongly implicated in the development of tissue infarction in response to focal cerebral ischemia. Nitric oxide and its derivatives can inhibit components of the electron transport chain, providing a likely target for these substances in ischemic and post-ischemic brain. Lactate content is increased during post-ischemic reperfusion in tissue destined to become infarcted, consistent with impairment of mitochondrial respiration. To investigate the possible involvement of nitric oxide in generating these changes, we have tested the effect of 7-nitroindazole, a nitric oxide synthase (NOS) inhibitor, on the content of lactate and other metabolites during early reperfusion following temporary focal ischemia. This treatment inhibited total NOS by approximately 50%. However, the treatment did not significantly affect the marked increases in lactate in post-ischemic brain nor did it alter the recovery of other energy-related metabolites. These findings indicate that inhibition of oxidative metabolism is probably not the primary site of the deleterious effects of nitric oxide and derivatives during early post-ischemic reperfusion.


Subject(s)
Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Ischemic Attack, Transient/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glucose/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Infarction, Middle Cerebral Artery/surgery , Lactic Acid/metabolism , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Phosphocreatine/metabolism , Rats , Rats, Sprague-Dawley
20.
J Neurochem ; 102(4): 1369-82, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17484727

ABSTRACT

The major cellular antioxidant, glutathione, is mostly localized in the cytosol but a small portion is found in mitochondria. We have recently shown that highly selective depletion of mitochondrial glutathione in astrocytes in culture markedly increased cell death induced by the peroxynitrite donor, 3-morpholino-syndnonimine. The present study was aimed at characterizing the increase in susceptibility arising from mitochondrial glutathione loss and testing the possibility that elevating this metabolite pool above normal values could be protective. The increased vulnerability of astrocytes with depleted mitochondrial glutathione to Sin-1 was confirmed. Furthermore, these cells showed marked increases in sensitivity to hydrogen peroxide and also to high concentrations of the nitric oxide donor, S-nitroso-N-acetyl-penicillamine. The increase in cell death was mostly due to necrosis as indicated by substantially increased release of lactate dehydrogenase and staining of nuclei with propidium iodide but little change in annexin V staining and caspase 3 activation. The enhanced cell loss was blocked by prior restoration of the mitochondrial glutathione content. It was also essentially fully inhibited by treatment with cyclosporin A, consistent with a role for the mitochondrial permeability transition in the development of cell death. Susceptibility to the classical apoptosis inducer, staurosporine, was only affected to a small extent in contrast to the response to the other substances tested. Incubation of normal astrocytes with glutathione monoethylester produced large and long-lasting increases in mitochondrial glutathione content with much smaller effects on the cytosolic glutathione pool. This treatment reduced cell death on exposure to 3-morpholino-syndnonimine or hydrogen peroxide but not S-nitroso-N-acetyl-pencillamine or staurosporine. These findings provide evidence for an important role for mitochondrial glutathione in preserving cell viability during periods of oxidative or nitrative stress and indicate that increases in this glutathione pool can confer protection against some of these stressors.


Subject(s)
Astrocytes/metabolism , Cell Death/physiology , Glutathione/metabolism , Mitochondria/metabolism , Nitrates/metabolism , Oxidative Stress/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/ultrastructure , Caspase 3/metabolism , Cell Death/drug effects , Cells, Cultured , Citrate (si)-Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , L-Lactate Dehydrogenase/metabolism , Mitochondria/drug effects , Oxidants/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL