1
|
Dumanska H, Telka M, Veselovsky N. Inhibition of high-voltage-activated calcium currents by acute hypoxia in cultured retinal ganglion cells. Front Cell Neurosci 2023; 17:1202083. [PMID: 37465211 PMCID: PMC10351036 DOI: 10.3389/fncel.2023.1202083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Hypoxia is a common factor of numerous ocular diseases that lead to dysfunctions and loss of retinal ganglion cells (RGCs) with subsequent vision loss. High-voltage-activated calcium channels are the main source of calcium entry into neurons. Their activity plays a central role in different signaling processes in health and diseases, such as enzyme activation, gene transcription, synaptic transmission, or the onset of cell death. This study aims to establish and evaluate the initial effect of the early stage of acute hypoxia on somatic HVA calcium currents in cultured RGCs. HVA calcium currents were recorded in RGCs using the whole-cell patch-clamp technique in the voltage-clamp mode. The fast local superfusion was used for a brief (up to 270 s) application of the hypoxic solution (pO2 < 5 mmHg). The switch from normoxic to hypoxic solutions and vice versa was less than 1 s. The HVA calcium channel activity was inhibited by acute hypoxia in 79% of RGCs (30 of 38 RGCs) in a strong voltage-dependent manner. The level of inhibition was independent of the duration of hypoxia or repeated applications. The hypoxia-induced inhibition of calcium currents had a strong correlation with the duration of hypoxia and showed the transition from reversible to irreversible at 75 s of hypoxia and longer. The results obtained are the first demonstration of the phenomena of HVA calcium current inhibition by acute hypoxia in RGCs and provide a conceptual framework for further research.
Collapse
|
2
|
Escobar I, Xu J, Jackson CW, Stegelmann SD, Fagerli EA, Dave KR, Perez-Pinzon MA. Resveratrol Preconditioning Protects Against Ischemia-Induced Synaptic Dysfunction and Cofilin Hyperactivation in the Mouse Hippocampal Slice. Neurotherapeutics 2023; 20:1177-1197. [PMID: 37208551 PMCID: PMC10457274 DOI: 10.1007/s13311-023-01386-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/21/2023] Open
Abstract
Perturbations in synaptic function are major determinants of several neurological diseases and have been associated with cognitive impairments after cerebral ischemia (CI). Although the mechanisms underlying CI-induced synaptic dysfunction have not been well defined, evidence suggests that early hyperactivation of the actin-binding protein, cofilin, plays a role. Given that synaptic impairments manifest shortly after CI, prophylactic strategies may offer a better approach to prevent/mitigate synaptic damage following an ischemic event. Our laboratory has previously demonstrated that resveratrol preconditioning (RPC) promotes cerebral ischemic tolerance, with many groups highlighting beneficial effects of resveratrol treatment on synaptic and cognitive function in other neurological conditions. Herein, we hypothesized that RPC would mitigate hippocampal synaptic dysfunction and pathological cofilin hyperactivation in an ex vivo model of ischemia. Various electrophysiological parameters and synaptic-related protein expression changes were measured under normal and ischemic conditions utilizing acute hippocampal slices derived from adult male mice treated with resveratrol (10 mg/kg) or vehicle 48 h prior. Remarkably, RPC significantly increased the latency to anoxic depolarization, decreased cytosolic calcium accumulation, prevented aberrant increases in synaptic transmission, and rescued deficits in long-term potentiation following ischemia. Additionally, RPC upregulated the expression of the activity-regulated cytoskeleton associated protein, Arc, which was partially required for RPC-mediated attenuation of cofilin hyperactivation. Taken together, these findings support a role for RPC in mitigating CI-induced excitotoxicity, synaptic dysfunction, and pathological over-activation of cofilin. Our study provides further insight into mechanisms underlying RPC-mediated neuroprotection against CI and implicates RPC as a promising strategy to preserve synaptic function after ischemia.
Collapse
Affiliation(s)
- Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Jing Xu
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Charles W Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Samuel D Stegelmann
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Eric A Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
3
|
Abstract
Disruptions in the delivery of oxygen and glucose impair the function of neural circuits, with lethal consequences commonly observed in stroke and cardiac arrest. Intense focus has been placed on understanding how to overcome neuronal failure during energy stress. Important insights into neuroprotective strategies have come from studies of evolutionary adaptations for survival in hypoxic environments, such as those seen in turtles, naked mole-rats, and several other animals1. Amphibians are not usually numbered among 'champion' hypoxia-tolerant vertebrates, yet here we demonstrate a massive increase in the capacity of a neural circuit to produce activity following oxygen and glucose deprivation in adult bullfrogs. Rhythmic output from a brainstem circuit failed following minutes of severe hypoxia and simulated ischemia; however, after hibernation this network produced patterned activity for ∼3.5 hours during severe hypoxia and ∼2 hours in ischemia. This remarkable improvement was supported by a switch to brain glycogen to fuel anaerobic glycolysis, a pathway thought to support neuronal homeostasis for only a few minutes during ischemia2. These results reveal that circuit activity can exhibit dramatic metabolic plasticity that minimizes the need for ATP synthesis, and these findings represent the greatest range in hypoxia tolerance within a vertebrate neural network. Uncovering the rules that allow the brain to flexibly run only on endogenous fuel reserves will reveal new insights into brain energetics, circuit evolution, and neuroprotection.
Collapse
|
4
|
Energy matters: presynaptic metabolism and the maintenance of synaptic transmission. Nat Rev Neurosci 2021; 23:4-22. [PMID: 34782781 DOI: 10.1038/s41583-021-00535-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
Abstract
Synaptic activity imposes large energy demands that are met by local adenosine triphosphate (ATP) synthesis through glycolysis and mitochondrial oxidative phosphorylation. ATP drives action potentials, supports synapse assembly and remodelling, and fuels synaptic vesicle filling and recycling, thus sustaining synaptic transmission. Given their polarized morphological features - including long axons and extensive branching in their terminal regions - neurons face exceptional challenges in maintaining presynaptic energy homeostasis, particularly during intensive synaptic activity. Recent studies have started to uncover the mechanisms and signalling pathways involved in activity-dependent and energy-sensitive regulation of presynaptic energetics, or 'synaptoenergetics'. These conceptual advances have established the energetic regulation of synaptic efficacy and plasticity as an exciting research field that is relevant to a range of neurological disorders associated with bioenergetic failure and synaptic dysfunction.
Collapse
|
5
|
Astrocyte Gliotransmission in the Regulation of Systemic Metabolism. Metabolites 2021; 11:metabo11110732. [PMID: 34822390 PMCID: PMC8623475 DOI: 10.3390/metabo11110732] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
Normal brain function highly relies on the appropriate functioning of astrocytes. These glial cells are strategically situated between blood vessels and neurons, provide significant substrate support to neuronal demand, and are sensitive to neuronal activity and energy-related molecules. Astrocytes respond to many metabolic conditions and regulate a wide array of physiological processes, including cerebral vascular remodeling, glucose sensing, feeding, and circadian rhythms for the control of systemic metabolism and behavior-related responses. This regulation ultimately elicits counterregulatory mechanisms in order to couple whole-body energy availability with brain function. Therefore, understanding the role of astrocyte crosstalk with neighboring cells via the release of molecules, e.g., gliotransmitters, into the parenchyma in response to metabolic and neuronal cues is of fundamental relevance to elucidate the distinct roles of these glial cells in the neuroendocrine control of metabolism. Here, we review the mechanisms underlying astrocyte-released gliotransmitters that have been reported to be crucial for maintaining homeostatic regulation of systemic metabolism.
Collapse
|
6
|
Adams S, Zubov T, Bueschke N, Santin JM. Neuromodulation or energy failure? Metabolic limitations silence network output in the hypoxic amphibian brainstem. Am J Physiol Regul Integr Comp Physiol 2021; 320:R105-R116. [PMID: 33175586 PMCID: PMC7948128 DOI: 10.1152/ajpregu.00209.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
Hypoxia tolerance in the vertebrate brain often involves chemical modulators that arrest neuronal activity to conserve energy. However, in intact networks, it can be difficult to determine whether hypoxia triggers modulators to stop activity in a protective manner or whether activity stops because rates of ATP synthesis are insufficient to support network function. Here, we assessed the extent to which neuromodulation or metabolic limitations arrest activity in the respiratory network of bullfrogs-a circuit that survives moderate periods of oxygen deprivation, presumably, by activating an inhibitory noradrenergic pathway. We confirmed that hypoxia and norepinephrine (NE) reduce network output, consistent with the view that hypoxia may cause the release of NE to inhibit activity. However, these responses differed qualitatively; hypoxia, but not NE, elicited a large motor burst and silenced the network. The stereotyped response to hypoxia persisted in the presence of both NE and an adrenergic receptor blocker that eliminates sensitivity to NE, indicating that noradrenergic signaling does not cause the arrest. Pharmacological inhibition of glycolysis and mitochondrial respiration recapitulated all features of hypoxia on network activity, implying that reduced ATP synthesis underlies the effects of hypoxia. Finally, activating modulatory mechanisms that dampen neuronal excitability when ATP levels fall, KATP channels and AMP-dependent protein kinase, did not resemble the hypoxic response. These results suggest that energy failure-rather than inhibitory modulation-silences the respiratory network during hypoxia and emphasize the need to account for metabolic limitations before concluding that modulators arrest activity as an adaptation for energy conservation in the nervous system.
Collapse
Affiliation(s)
- Sasha Adams
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Tanya Zubov
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Nikolaus Bueschke
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Joseph M Santin
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| |
Collapse
|
7
|
Andreasen M, Nedergaard S. Effect of acute mitochondrial dysfunction on hyperexcitable network activity in rat hippocampus in vitro. Brain Res 2020; 1751:147193. [PMID: 33157100 DOI: 10.1016/j.brainres.2020.147193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 11/15/2022]
Abstract
Metabolic stress imposed by epileptic seizures can result in mitochondrial dysfunction, believed to act as positive feedback on epileptogenesis and seizure susceptibility. As the mechanism behind this positive feedback is unclear, the aim of the present study was to investigate the causal link between acute mitochondrial dysfunction and increased seizure susceptibility in hyperexcitable hippocampal networks. Following the induction of spontaneous interictal-like discharges, acute selective pharmacological blockade of either of the mitochondrial respiratory complexes (MRC) I-IV induced seizure-like events (SLE) in 78-100% of experiments. A similar result was obtained by uncoupling the oxidative phosphorylation (OXPHOS) but not by selective blockade of MRCV (ATP synthase) which did not induce SLE. The reactive oxygen species (ROS) scavenger 4-hydroxy-2,2,6,6-tetramethylpiperidine 1-oxyl (tempol, 2 mM) significantly reduced the proconvulsant effect of blocking MRCI but did not reduce the proconvulsant effect of OXPHOS uncoupling. These findings indicate that acute mitochondrial dysfunction can lead to a convulsive state within a short timeframe, and that increased ROS production makes substantial contribution to such induction in addition to other mitochondrial related factors, which appears to be independent of changes in ROS and ATP production.
Collapse
Affiliation(s)
- Mogens Andreasen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.
| | - Steen Nedergaard
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
8
|
Tanaka Y, Nakagomi N, Doe N, Nakano-Doi A, Sawano T, Takagi T, Matsuyama T, Yoshimura S, Nakagomi T. Early Reperfusion Following Ischemic Stroke Provides Beneficial Effects, Even After Lethal Ischemia with Mature Neural Cell Death. Cells 2020; 9:cells9061374. [PMID: 32492968 PMCID: PMC7349270 DOI: 10.3390/cells9061374] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is a critical disease caused by cerebral artery occlusion in the central nervous system (CNS). Recent therapeutic advances, such as neuroendovascular intervention and thrombolytic therapy, have allowed recanalization of occluded brain arteries in an increasing number of stroke patients. Although previous studies have focused on rescuing neural cells that still survive despite decreased blood flow, expanding the therapeutic time window may allow more patients to undergo reperfusion in the near future, even after lethal ischemia, which is characterized by death of mature neural cells, such as neurons and glia. However, it remains unclear whether early reperfusion following lethal ischemia results in positive outcomes. The present study used two ischemic mouse models—90-min transient middle cerebral artery occlusion (t-MCAO) paired with reperfusion to induce lethal ischemia and permanent middle cerebral artery occlusion (p-MCAO)—to investigate the effect of early reperfusion up to 8 w following MCAO. Although early reperfusion following 90-min t-MCAO did not rescue mature neural cells, it preserved the vascular cells within the ischemic areas at 1 d following 90-min t-MCAO compared to that following p-MCAO. In addition, early reperfusion facilitated the healing processes, including not only vascular but also neural repair, during acute and chronic periods and improved recovery. Furthermore, compared with p-MCAO, early reperfusion after t-MCAO prevented behavioral symptoms of neurological deficits without increasing negative complications, including hemorrhagic transformation and mortality. These results indicate that early reperfusion provides beneficial effects presumably via cytoprotective and regenerative mechanisms in the CNS, suggesting that it may be useful for stroke patients that experienced lethal ischemia.
Collapse
Affiliation(s)
- Yasue Tanaka
- Department of Neurosurgery, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan; (Y.T.); (T.T.)
| | - Nami Nakagomi
- Department of Surgical Pathology, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
| | - Nobutaka Doe
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan;
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
| | - Toshinori Sawano
- Department of Biomedical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan;
| | - Toshinori Takagi
- Department of Neurosurgery, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan; (Y.T.); (T.T.)
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan; (Y.T.); (T.T.)
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
- Correspondence: (S.Y.); (T.N.); Tel.: +81-798-45-6455 (S.Y.); +81-798-45-6821 (T.N.)
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan;
- Correspondence: (S.Y.); (T.N.); Tel.: +81-798-45-6455 (S.Y.); +81-798-45-6821 (T.N.)
| |
Collapse
|
9
|
Flavin Adenine Dinucleotide Fluorescence as an Early Marker of Mitochondrial Impairment During Brain Hypoxia. Int J Mol Sci 2020; 21:ijms21113977. [PMID: 32492921 PMCID: PMC7312830 DOI: 10.3390/ijms21113977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/28/2020] [Accepted: 05/30/2020] [Indexed: 12/31/2022] Open
Abstract
Multimodal continuous bedside monitoring is increasingly recognized as a promising option for early treatment stratification in patients at risk for ischemia during neurocritical care. Modalities used at present are, for example, oxygen availability and subdural electrocorticography. The assessment of mitochondrial function could be an interesting complement to these modalities. For instance, flavin adenine dinucleotide (FAD) fluorescence permits direct insight into the mitochondrial redox state. Therefore, we explored the possibility of using FAD fluorometry to monitor consequences of hypoxia in brain tissue in vitro and in vivo. By combining experimental results with computational modeling, we identified the potential source responsible for the fluorescence signal and gained insight into the hypoxia-associated metabolic changes in neuronal energy metabolism. In vitro, hypoxia was characterized by a reductive shift of FAD, impairment of synaptic transmission and increasing interstitial potassium [K+]o. Computer simulations predicted FAD changes to originate from the citric acid cycle enzyme α-ketoglutarate dehydrogenase and pyruvate dehydrogenase. In vivo, the FAD signal during early hypoxia displayed a reductive shift followed by a short oxidation associated with terminal spreading depolarization. In silico, initial tissue hypoxia followed by a transient re-oxygenation phase due to glucose depletion might explain FAD dynamics in vivo. Our work suggests that FAD fluorescence could be readily used to monitor mitochondrial function during hypoxia and represents a potential diagnostic tool to differentiate underlying metabolic processes for complementation of multimodal brain monitoring.
Collapse
|
10
|
Mild hypothermia protects synaptic transmission from experimental ischemia through reduction in the function of nucleoside transporters in the mouse hippocampus. Neuropharmacology 2019; 163:107853. [PMID: 31734385 DOI: 10.1016/j.neuropharm.2019.107853] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/28/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022]
Abstract
Ischemia, a severe metabolic stress, increases adenosine levels and causes the suppression of synaptic transmission through adenosine A1 receptors. Although temperature also regulates extracellular adenosine levels, the effect of temperature on ischemia-induced activation of adenosine receptors is not yet fully understood. Here we examined the role of adenosine A1 receptors in mild hypothermia-mediated neuroprotection during the acute phase of ischemia. Severe ischemia-induced neurosynaptic impairment was reproduced by oxygen-glucose deprivation at normothermia (36 °C) and assessed with extracellular recordings or whole-cell patch clamp recordings in acute hippocampal slices in mice. Mild hypothermia (32 °C) induced the protection of synaptic transmission by activating adenosine A1 receptors. Stricter hypothermia (28 °C) caused additional neuroprotective effects by extending the onset time to anoxic depolarization; however, this effect was not associated with adenosine A1 receptors. The response of exogenous adenosine-induced inhibition of hippocampal synaptic transmission was increased by lowering the temperature to 32 °C or 28 °C. Hypothermia also reduced the function of dipryidamole-sensitive nucleoside transporters. These findings suggest that an increased response of adenosine A1 receptors, caused by a reduction in the function of nucleoside transporters, is one mechanism by which therapeutic hypothermia (usually used within the mild range) mediates neurosynaptic protection in the acute phase of stroke.
Collapse
|
11
|
Chinopoulos C, Seyfried TN. Mitochondrial Substrate-Level Phosphorylation as Energy Source for Glioblastoma: Review and Hypothesis. ASN Neuro 2019; 10:1759091418818261. [PMID: 30909720 PMCID: PMC6311572 DOI: 10.1177/1759091418818261] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant of the primary adult brain cancers. Ultrastructural and biochemical evidence shows that GBM cells exhibit mitochondrial abnormalities incompatible with energy production through oxidative phosphorylation (OxPhos). Under such conditions, the mitochondrial F0-F1 ATP synthase operates in reverse at the expense of ATP hydrolysis to maintain a moderate membrane potential. Moreover, expression of the dimeric M2 isoform of pyruvate kinase in GBM results in diminished ATP output, precluding a significant ATP production from glycolysis. If ATP synthesis through both glycolysis and OxPhos was impeded, then where would GBM cells obtain high-energy phosphates for growth and invasion? Literature is reviewed suggesting that the succinate-CoA ligase reaction in the tricarboxylic acid cycle can substantiate sufficient ATP through mitochondrial substrate-level phosphorylation (mSLP) to maintain GBM growth when OxPhos is impaired. Production of high-energy phosphates would be supported by glutaminolysis—a hallmark of GBM metabolism—through the sequential conversion of glutamine → glutamate → alpha-ketoglutarate → succinyl CoA → succinate. Equally important, provision of ATP through mSLP would maintain the adenine nucleotide translocase in forward mode, thus preventing the reverse-operating F0-F1 ATP synthase from depleting cytosolic ATP reserves. Because glucose and glutamine are the primary fuels driving the rapid growth of GBM and most tumors for that matter, simultaneous restriction of these two substrates or inhibition of mSLP should diminish cancer viability, growth, and invasion.
Collapse
|
12
|
Povysheva N, Nigam A, Brisbin AK, Johnson JW, Barrionuevo G. Oxygen-Glucose Deprivation Differentially Affects Neocortical Pyramidal Neurons and Parvalbumin-Positive Interneurons. Neuroscience 2019; 412:72-82. [PMID: 31152933 DOI: 10.1016/j.neuroscience.2019.05.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 05/02/2019] [Accepted: 05/21/2019] [Indexed: 11/28/2022]
Abstract
Stroke is a devastating brain disorder. The pathophysiology of stroke is associated with an impaired excitation-inhibition balance in the area that surrounds the infarct core after the insult, the peri-infarct zone. Here we exposed slices from adult mouse prefrontal cortex to oxygen-glucose deprivation and reoxygenation (OGD-RO) to study ischemia-induced changes in the activity of excitatory pyramidal neurons and inhibitory parvalbumin (PV)-positive interneurons. We found that during current-clamp recordings, PV-positive interneurons were more vulnerable to OGD-RO than pyramidal neurons as indicated by the lower percentage of recovery of PV-positive interneurons. However, neither the amplitude of OGD-induced depolarization observed in current-clamp mode nor the OGD-associated current observed in voltage-clamp mode differed between the two cell types. Large amplitude, presumably action-potential dependent, spontaneous postsynaptic inhibitory currents recorded from pyramidal neurons were less frequent after OGD-RO than in control condition. Disynaptic inhibitory postsynaptic currents (dIPSCs) in pyramidal neurons produced predominantly by PV-positive interneurons were reduced by OGD-RO. Following OGD-RO, dendrites of PV-positive interneurons exhibited more pathological beading than those of pyramidal neurons. Our data support the hypothesis that the differential vulnerability to ischemia-like conditions of excitatory and inhibitory neurons leads to the altered excitation-inhibition balance associated with stroke pathophysiology.
Collapse
Affiliation(s)
- Nadya Povysheva
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| | - Aparna Nigam
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Alyssa K Brisbin
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Germán Barrionuevo
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
13
|
LaCorte S. How chronic administration of benzodiazepines leads to unexplained chronic illnesses: A hypothesis. Med Hypotheses 2018; 118:59-67. [PMID: 30037616 DOI: 10.1016/j.mehy.2018.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/28/2018] [Accepted: 06/19/2018] [Indexed: 11/15/2022]
Abstract
It is thought that an ill defined biochemical cascade may lead to protracted withdrawal symptoms subsequent to discontinuance of routine use of benzodiazepine class drugs and establish chronic illness in some patients. In this review, published findings are presented that support the novel concept that withdrawal from benzodiazepine class drugs can trigger elevated and sustained levels of a potent oxidant called peroxynitrite via potentiation of the L-type voltage-gated calcium channels, and in the later stages of withdrawal, via excessive N-methyl-D-aspartate receptor activity, as well. Potentiation of L-type voltage-gated calcium channels and excessive N-methyl-D-aspartate receptor activity both result in calcium influx into the cell that triggers nitric oxide synthesis. In pathophysiological conditions, such increased nitric oxide synthesis leads to peroxynitrite formation. The downstream effects of peroxynitrite formation that may occur during withdrawal ultimately lead to further peroxynitrite production in a system of overlapping vicious cycles collectively referred to as the NO/ONOO(-) cycle. Once triggered, the elements of the NO/ONOO(-) cycle perpetuate pathophysiology, perhaps including reduced GABAA receptor functioning, that may explain protracted withdrawal associated symptoms while the vicious cycle nature of the NO/ONOO(-) cycle may explain how withdrawal becomes a chronic state. Suboptimal levels of tetrahydrobiopterin may be one risk factor for the development of the protracted withdrawal syndrome as this will lead to partial nitric oxide uncoupling and resultant peroxynitrite formation. Nitric oxide uncoupling results in superoxide production as calcium-dependent nitric oxide synthases attempt to produce nitric oxide in response to L-type voltage-gated calcium channel-mediated calcium influx that is known to occur during withdrawal. The combination of nitric oxide and superoxide produced, as when partial uncoupling occurs, react together in a very rapid, diffusion limited reaction to form peroxynitrite and thereby trigger the NO/ONOO(-) cycle. The NO/ONOO(-) cycle may explain the nature of the protracted withdrawal syndrome and the related constellation of symptoms that are also common in other illnesses characterized as NO/ONOO(-) disorders such as myalgic encephalomyelitis/chronic fatigue syndrome and fibromyalgia.
Collapse
Affiliation(s)
- S LaCorte
- Benzodiazepine Information Coalition, 1042 Fort Union Blvd. Suite 1030, Midvale, UT 84047, United States.
| |
Collapse
|
14
|
Rich LR, Brown AM. Fibre sub-type specific conduction reveals metabolic function in mouse sciatic nerve. J Physiol 2018. [PMID: 29517809 DOI: 10.1113/jp275680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We have developed an improved method that enables simultaneous recording of stimulus evoked compound action potentials from large myelinated A fibres and small unmyelinated C fibres in mouse sciatic nerves. Investigations into the ability of fructose to support conduction in sciatic nerve revealed a novel glia-to-axon metabolic pathway in which fructose is converted in Schwann cells to lactate for subsequent shuttling to A fibres. The C fibres most likely directly take up and metabolise fructose. These differences are indicative of fibre sub-type specific metabolic profiles. These results demonstrate that the physiological insights provided by the method can be applied to investigations of peripheral nerve, with a view to understanding the metabolic disruptions that underlie diabetic neuropathy. ABSTRACT The stimulus evoked compound action potential (CAP), recorded using suction electrodes, provides an index of the relative number of conducting axons within a nerve trunk. As such the CAP has been used to elucidate the diverse mechanisms of injury resulting from a variety of metabolic insults to central nervous white matter, whilst also providing a model with which to assess the benefits of clinically relevant neuroprotective strategies. In addition the technique lends itself to the study of metabolic cell-to-cell signalling that occurs between glial cells and neurones, and to exploring the ability of non-glucose substrates to support axon conduction. Although peripheral nerves are sensitive to metabolic insult and are susceptible to diabetic neuropathy, there is a lack of fundamental information regarding peripheral nerve metabolism. A confounding factor in such studies is the extended duration demanded by the experimental protocol, requiring stable recording for periods of many hours. We describe a method that allows us to record simultaneously the stimulus evoked CAPs from A and C fibres from mouse sciatic nerve, and demonstrate its utility as applied to investigations into fibre sub-type substrate use. Our results suggest that C fibres directly take up and metabolise fructose, whereas A fibre conduction is supported by fructose-derived lactate, implying there exist unique metabolic profiles in neighbouring fibre sub-types present within the same nerve trunk.
Collapse
Affiliation(s)
- Laura R Rich
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Angus M Brown
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.,Department of Neurology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
15
|
Du Y, Wang W, Lutton AD, Kiyoshi CM, Ma B, Taylor AT, Olesik JW, McTigue DM, Askwith CC, Zhou M. Dissipation of transmembrane potassium gradient is the main cause of cerebral ischemia-induced depolarization in astrocytes and neurons. Exp Neurol 2018; 303:1-11. [PMID: 29407729 DOI: 10.1016/j.expneurol.2018.01.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/02/2018] [Accepted: 01/25/2018] [Indexed: 01/16/2023]
Abstract
Membrane potential (VM) depolarization occurs immediately following cerebral ischemia and is devastating for the astrocyte homeostasis and neuronal signaling. Previously, an excessive release of extracellular K+ and glutamate has been shown to underlie an ischemia-induced VM depolarization. Ischemic insults should impair membrane ion channels and disrupt the physiological ion gradients. However, their respective contribution to ischemia-induced neuronal and glial depolarization and loss of neuronal excitability are unanswered questions. A short-term oxygen-glucose deprivation (OGD) was used for the purpose of examining the acute effect of ischemic conditions on ion channel activity and physiological K+ gradient in neurons and glial cells. We show that a 30 min OGD treatment exerted no measurable damage to the function of membrane ion channels in neurons, astrocytes, and NG2 glia. As a result of the resilience of membrane ion channels, neuronal spikes last twice as long as our previously reported 15 min time window. In the electrophysiological analysis, a 30 min OGD-induced dissipation of transmembrane K+ gradient contributed differently in brain cell depolarization: severe in astrocytes and neurons, and undetectable in NG2 glia. The discrete cellular responses to OGD corresponded to a total loss of 69% of the intracellular K+ contents in hippocampal slices as measured by Inductively Coupled Plasma Mass Spectrometry (ICP-MS). A major brain cell depolarization mechanism identified here is important for our understanding of cerebral ischemia pathology. Additionally, further understanding of the resilient response of NG2 glia to ischemia-induced intracellular K+ loss and depolarization should facilitate the development of future stroke therapy.
Collapse
Affiliation(s)
- Yixing Du
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei Wang
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anthony D Lutton
- Trace Element Research Laboratory, The Ohio State University, Columbus, OH 43210, USA
| | - Conrad M Kiyoshi
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Baofeng Ma
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anne T Taylor
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - John W Olesik
- Trace Element Research Laboratory, The Ohio State University, Columbus, OH 43210, USA
| | - Dana M McTigue
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Candice C Askwith
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Min Zhou
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
16
|
Cohan CH, Stradecki-Cohan HM, Morris-Blanco KC, Khoury N, Koronowski KB, Youbi M, Wright CB, Perez-Pinzon MA. Protein kinase C epsilon delays latency until anoxic depolarization through arc expression and GluR2 internalization. J Cereb Blood Flow Metab 2017; 37:3774-3788. [PMID: 28585865 PMCID: PMC5718329 DOI: 10.1177/0271678x17712178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Global cerebral ischemia is a debilitating injury that damages the CA1 region of the hippocampus, an area important for learning and memory. Protein kinase C epsilon (PKCɛ) activation is a critical component of many neuroprotective treatments. The ability of PKCɛ activation to regulate AMPA receptors (AMPARs) remains unexplored despite the role of AMPARs in excitotoxicity after brain ischemia. We determined that PKCɛ activation increased expression of a protein linked to learning and memory, activity-regulated cytoskeleton-associated protein (arc). Also, arc is necessary for neuroprotection and confers protection through decreasing AMPAR currents via GluR2 internalization. In vivo, activation of PKCɛ increased arc expression through a BDNF/TrkB pathway, and decreased GluR2 mRNA levels. In hippocampal cultured slices, PKCɛ activation decreased AMPAR current amplitudes in an arc- and GluR2-dependent manner. Additionally, PKCɛ activation triggered an arc- and GluR2 internalization-dependent delay in latency until anoxic depolarization. Inhibiting arc also blocked PKCɛ-mediated neuroprotection against lethal oxygen and glucose deprivation. These data characterize a novel PKCɛ-dependent mechanism that for the first time defines a role for arc and AMPAR internalization in conferring neuroprotection.
Collapse
Affiliation(s)
- Charles H Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Holly M Stradecki-Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kahlilia C Morris-Blanco
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Nathalie Khoury
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kevin B Koronowski
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Mehdi Youbi
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Clinton B Wright
- 2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Nortley R, Attwell D. Control of brain energy supply by astrocytes. Curr Opin Neurobiol 2017; 47:80-85. [PMID: 29054039 DOI: 10.1016/j.conb.2017.09.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 01/03/2023]
Abstract
Astrocytes form an anatomical bridge between the vasculature and neuronal synapses. Recent work suggests that they play a key role in regulating brain energy supply by increasing blood flow to regions where neurons are active, and setting the baseline level of blood flow. Controversy persists over whether lactate derived from astrocyte glycolysis is used to power oxidative phosphorylation in neurons, but astrocytes sustain neuronal ATP production by recycling neurotransmitter glutamate that would otherwise need to be resynthesised from glucose, and by providing a short-term energy store in the form of glycogen that can be mobilised when neurons are active.
Collapse
Affiliation(s)
- Ross Nortley
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
18
|
Mishchenko EL, Petrovskaya OV, Mishchenko AM, Petrovskiy ED, Ivanisenko NV, Ivanisenko VA. Integrated mathematical models for describing complex biological processes. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s0006350917050141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
19
|
Wang X, Ma Z, Fu Z, Gao S, Yang L, Jin Y, Sun H, Wang C, Fan W, Chen L, Zheng QY, Bi G, Ma CL. Hydroxysafflor Yellow A Protects Neurons From Excitotoxic Death through Inhibition of NMDARs. ASN Neuro 2016; 8:8/2/1759091416642345. [PMID: 27067428 PMCID: PMC4828664 DOI: 10.1177/1759091416642345] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 12/30/2015] [Indexed: 11/15/2022] Open
Abstract
Excessive glutamate release causes overactivation of N-methyl d-aspartate receptors (NMDARs), leading to excitatory neuronal damage in cerebral ischemia. Hydroxysafflor yellow A (HSYA), a compound extracted from Carthamus tinctorius L., has been reported to exert a neuroprotective effect in many pathological conditions, including brain ischemia. However, the underlying mechanism of HSYA's effect on neurons remains elusive. In the present study, we conducted experiments using patch-clamp recording of mouse hippocampal slices. In addition, we performed Ca2+ imaging, Western blots, as well as mitochondrial-targeted circularly permuted yellow fluorescent protein transfection into cultured hippocampal neurons in order to decipher the physiological mechanism underlying HSYA's neuroprotective effect. Through the electrophysiology experiments, we found that HSYA inhibited NMDAR-mediated excitatory postsynaptic currents without affecting α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor and γ-aminobutyric acid A-type receptor-mediated currents. This inhibitory effect of HSYA on NMDARs was concentration dependent. HSYA did not show any preferential inhibition of either N-methyl d-aspartate receptor subtype 2A- or N-methyl d-aspartate receptor subtype 2B- subunit-containing NMDARs. Additionally, HSYA exhibits a facilitatory effect on paired NMDAR-mediated excitatory postsynaptic currents. Furthermore, HSYA reduced the magnitude of NMDAR-mediated membrane depolarization currents evoked by oxygen-glucose deprivation, and suppressed oxygen-glucose deprivation–induced and NMDAR-dependent ischemic long-term potentiation, which is believed to cause severe reperfusion damage after ischemia. Through the molecular biology experiments, we found that HSYA inhibited the NMDA-induced and NMDAR-mediated intracellular Ca2+ concentration increase in hippocampal cultures, reduced apoptotic and necrotic cell deaths, and prevented mitochondrial damage. Together, our data demonstrate for the first time that HSYA protects hippocampal neurons from excitotoxic damage through the inhibition of NMDARs. This novel finding indicates that HSYA may be a promising pharmacological candidate for the treatment of brain ischemia.
Collapse
Affiliation(s)
- Xingtao Wang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China "Brain stroke" Key Lab of Shandong Health Administration Institute, Binzhou Medical University, Yantai, Shandong, China Department of Internal Neurology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Zhiyuan Ma
- School of Public Economics and Administration, Shanghai University of Finance and Economics, Shanghai, China
| | - Zhongxiao Fu
- CAS Key Laboratory of Brain Function and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Su Gao
- Department of Internal Neurology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Liu Yang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China "Brain stroke" Key Lab of Shandong Health Administration Institute, Binzhou Medical University, Yantai, Shandong, China
| | - Yan Jin
- CAS Key Laboratory of Brain Function and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Sun
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China "Brain stroke" Key Lab of Shandong Health Administration Institute, Binzhou Medical University, Yantai, Shandong, China
| | - Chaoyun Wang
- Department of Pharmacology, Binzhou Medical University, Yantai, Shandong, China
| | - Weiming Fan
- Department of Internal Neurology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Lin Chen
- CAS Key Laboratory of Brain Function and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Qing-Yin Zheng
- Department of Internal Neurology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Guoqiang Bi
- CAS Key Laboratory of Brain Function and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Chun-Lei Ma
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China "Brain stroke" Key Lab of Shandong Health Administration Institute, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
20
|
Hamilton NB, Kolodziejczyk K, Kougioumtzidou E, Attwell D. Proton-gated Ca(2+)-permeable TRP channels damage myelin in conditions mimicking ischaemia. Nature 2016; 529:523-7. [PMID: 26760212 PMCID: PMC4733665 DOI: 10.1038/nature16519] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/07/2015] [Indexed: 11/09/2022]
Abstract
The myelin sheaths wrapped around axons by oligodendrocytes are crucial for brain function. In ischaemia myelin is damaged in a Ca(2+)-dependent manner, abolishing action potential propagation. This has been attributed to glutamate release activating Ca(2+)-permeable N-methyl-D-aspartate (NMDA) receptors. Surprisingly, we now show that NMDA does not raise the intracellular Ca(2+) concentration ([Ca(2+)]i) in mature oligodendrocytes and that, although ischaemia evokes a glutamate-triggered membrane current, this is generated by a rise of extracellular [K(+)] and decrease of membrane K(+) conductance. Nevertheless, ischaemia raises oligodendrocyte [Ca(2+)]i, [Mg(2+)]i and [H(+)]i, and buffering intracellular pH reduces the [Ca(2+)]i and [Mg(2+)]i increases, showing that these are evoked by the rise of [H(+)]i. The H(+)-gated [Ca(2+)]i elevation is mediated by channels with characteristics of TRPA1, being inhibited by ruthenium red, isopentenyl pyrophosphate, HC-030031, A967079 or TRPA1 knockout. TRPA1 block reduces myelin damage in ischaemia. These data suggest that TRPA1-containing ion channels could be a therapeutic target in white matter ischaemia.
Collapse
Affiliation(s)
- Nicola B Hamilton
- Department of Neuroscience, Physiology &Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Karolina Kolodziejczyk
- Department of Neuroscience, Physiology &Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Eleni Kougioumtzidou
- Department of Neuroscience, Physiology &Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology &Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| |
Collapse
|
21
|
Abstract
Objective: To analyze the mechanism of neuroprotection of insulin and which blood glucose range was benefit for insulin exerting neuroprotective action. Data Sources: The study is based on the data from PubMed. Study Selection: Articles were selected with the search terms “insulin”, “blood glucose”, “neuroprotection”, “brain”, “glycogen”, “cerebral ischemia”, “neuronal necrosis”, “glutamate”, “γ-aminobutyric acid”. Results: Insulin has neuroprotection. The mechanisms include the regulation of neurotransmitter, promoting glycogen synthesis, and inhibition of neuronal necrosis and apoptosis. Insulin could play its role in neuroprotection by avoiding hypoglycemia and hyperglycemia. Conclusions: Intermittent and long-term infusion insulin may be a benefit for patients with ischemic brain damage at blood glucose 6–9 mmol/L.
Collapse
Affiliation(s)
| | - Yu Pei
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
22
|
|
23
|
Abstract
The term spreading depolarization (SD) refers to waves of abrupt, sustained mass depolarization in gray matter of the CNS. SD, which spreads from neuron to neuron in affected tissue, is characterized by a rapid near-breakdown of the neuronal transmembrane ion gradients. SD can be induced by hypoxic conditions--such as from ischemia--and facilitates neuronal death in energy-compromised tissue. SD has also been implicated in migraine aura, where SD is assumed to ascend in well-nourished tissue and is typically benign. In addition to these two ends of the "SD continuum," an SD wave can propagate from an energy-depleted tissue into surrounding, well-nourished tissue, as is often the case in stroke and brain trauma. This review presents the neurobiology of SD--its triggers and propagation mechanisms--as well as clinical manifestations of SD, including overlaps and differences between migraine aura and stroke, and recent developments in neuromonitoring aimed at better diagnosis and more targeted treatments.
Collapse
Affiliation(s)
- Jens P Dreier
- Department of Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany.
| | - Clemens Reiffurth
- Department of Experimental Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany; Center for Stroke Research, Charité University Medicine Berlin, 10117 Berlin, Germany
| |
Collapse
|
24
|
Abstract
Cancer is widely considered a genetic disease involving nuclear mutations in oncogenes and tumor suppressor genes. This view persists despite the numerous inconsistencies associated with the somatic mutation theory. In contrast to the somatic mutation theory, emerging evidence suggests that cancer is a mitochondrial metabolic disease, according to the original theory of Otto Warburg. The findings are reviewed from nuclear cytoplasm transfer experiments that relate to the origin of cancer. The evidence from these experiments is difficult to reconcile with the somatic mutation theory, but is consistent with the notion that cancer is primarily a mitochondrial metabolic disease.
Collapse
|
25
|
Völgyi K, Gulyássy P, Háden K, Kis V, Badics K, Kékesi KA, Simor A, Györffy B, Tóth EA, Lubec G, Juhász G, Dobolyi A. Synaptic mitochondria: a brain mitochondria cluster with a specific proteome. J Proteomics 2015; 120:142-57. [PMID: 25782751 DOI: 10.1016/j.jprot.2015.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/16/2015] [Accepted: 03/04/2015] [Indexed: 01/15/2023]
Abstract
UNLABELLED The synapse is a particularly important compartment of neurons. To reveal its molecular characteristics we isolated whole brain synaptic (sMito) and non-synaptic mitochondria (nsMito) from the mouse brain with purity validated by electron microscopy and fluorescence activated cell analysis and sorting. Two-dimensional differential gel electrophoresis and mass spectrometry based proteomics revealed 22 proteins with significantly higher and 34 proteins with significantly lower levels in sMito compared to nsMito. Expression differences in some oxidative stress related proteins, such as superoxide dismutase [Mn] (Sod2) and complement component 1Q subcomponent-binding protein (C1qbp), as well as some tricarboxylic acid cycle proteins, including isocitrate dehydrogenase subunit alpha (Idh3a) and ATP-forming β subunit of succinyl-CoA ligase (SuclA2), were verified by Western blot, the latter two also by immunohistochemistry. The data suggest altered tricarboxylic acid metabolism in energy supply of synapse while the marked differences in Sod2 and C1qbp support high sensitivity of synapses to oxidative stress. Further functional clustering demonstrated that proteins with higher synaptic levels are involved in synaptic transmission, lactate and glutathione metabolism. In contrast, mitochondrial proteins associated with glucose, lipid, ketone metabolism, signal transduction, morphogenesis, protein synthesis and transcription were enriched in nsMito. Altogether, the results suggest a specifically tuned composition of synaptic mitochondria. BIOLOGICAL SIGNIFICANCE Neurons communicate with each other through synapse, a compartment metabolically isolated from the cell body. Mitochondria are concentrated in presynaptic terminals by active transport to provide energy supply for information transfer. Mitochondrial composition in the synapse may be different than in the cell body as some examples have demonstrated altered mitochondrial composition with cell type and cellular function in the muscle, heart and liver. Therefore, we posed the question whether protein composition of synaptic mitochondria reflects its specific functions. The determined protein difference pattern was in accordance with known functional specialties of high demand synaptic mitochondria. The data also suggest specifically tuned metabolic fluxes for energy production by means of interaction with glial cells surrounding the synapse. These findings provide possible mechanisms for dynamically adapting synaptic mitochondrial output to actual demand. In turn, an increased vulnerability of synaptic mitochondria to oxidative stress is implied by the data. This is important from theoretical but potentially also from therapeutic aspects. Mitochondria are known to be affected in some neurodegenerative and psychiatric disorders, and proteins with elevated level in synaptic mitochondria, e.g. C1qbp represent targets for future drug development, by which synaptic and non-synaptic mitochondria can be differentially affected.
Collapse
Affiliation(s)
- Katalin Völgyi
- MTA-ELTE NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest H-1117, Hungary; Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Péter Gulyássy
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-TTK NAP MS Neuroproteomics Research Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | - Krisztina Háden
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Viktor Kis
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Kata Badics
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Katalin Adrienna Kékesi
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Attila Simor
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Balázs Györffy
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Eszter Angéla Tóth
- Department of Immunology, Eötvös Loránd University, Budapest H-1117, Hungary; Faculty of Science Research and Instrument Core Facility (ELTE FS-RICF), Eötvös Loránd University, Budapest H-1117, Hungary
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Vienna A-1090, Austria
| | - Gábor Juhász
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-TTK NAP MS Neuroproteomics Research Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | - Arpád Dobolyi
- MTA-ELTE NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest H-1117, Hungary.
| |
Collapse
|
26
|
Reactive oxygen species initiate a metabolic collapse in hippocampal slices: potential trigger of cortical spreading depression. J Cereb Blood Flow Metab 2014; 34:1540-9. [PMID: 25027308 PMCID: PMC4158675 DOI: 10.1038/jcbfm.2014.121] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/27/2014] [Accepted: 06/16/2014] [Indexed: 11/08/2022]
Abstract
Excessive accumulation of reactive oxygen species (ROS) underlies oxidative damage. We find that in hippocampal slices, decreased activity of glucose-based antioxidant system induces a massive, abrupt, and detrimental change in cellular functions. We call this phenomenon metabolic collapse (MC). This collapse manifested in long-lasting silencing of synaptic transmission, abnormal oxidation of NAD(P)H and FADH2 associated with immense oxygen consumption, and massive neuronal depolarization. MC occurred without any preceding deficiency in neuronal energy supply or disturbances of ionic homeostasis and spread throughout the hippocampus. It was associated with a preceding accumulation of ROS and was largely prevented by application of an efficient antioxidant Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl). The consequences of MC resemble cortical spreading depression (CSD), a wave of neuronal depolarization that occurs in migraine, brain trauma, and stroke, the cellular initiation mechanisms of which are poorly understood. We suggest that ROS accumulation might also be the primary trigger of CSD. Indeed, we found that Tempol strongly reduced occurrence of CSD in vivo, suggesting that ROS accumulation may be a key mechanism of CSD initiation.
Collapse
|
27
|
Pietrobon D, Moskowitz MA. Chaos and commotion in the wake of cortical spreading depression and spreading depolarizations. Nat Rev Neurosci 2014; 15:379-93. [PMID: 24857965 DOI: 10.1038/nrn3770] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Punctuated episodes of spreading depolarizations erupt in the brain, encumbering tissue structure and function, and raising fascinating unanswered questions concerning their initiation and propagation. Linked to migraine aura and headache, cortical spreading depression contributes to the morbidity in the world's migraine with aura population. Even more ominously, erupting spreading depolarizations accelerate tissue damage during brain injury. The once-held view that spreading depolarizations may not exist in the human brain has changed, largely because of the discovery of migraine genes that confer cortical spreading depression susceptibility, the application of sophisticated imaging tools and efforts to interrogate their impact in the acutely injured human brain.
Collapse
Affiliation(s)
- Daniela Pietrobon
- Department of Biomedical Sciences and CNR Institute of Neuroscience, University of Padova 35121 Padova, Italy
| | - Michael A Moskowitz
- 1] Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, 149 13th Street, Room 6403, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA. [2] Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, O'Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 2014; 508:55-60. [PMID: 24670647 PMCID: PMC3976267 DOI: 10.1038/nature13165] [Citation(s) in RCA: 1344] [Impact Index Per Article: 122.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 02/19/2014] [Indexed: 01/12/2023]
Abstract
Increases in brain blood flow, evoked by neuronal activity, power neural computation and form the basis of BOLD (blood-oxygen-level-dependent) functional imaging. Whether blood flow is controlled solely by arteriole smooth muscle, or also by capillary pericytes, is controversial. We demonstrate that neuronal activity and the neurotransmitter glutamate evoke the release of messengers that dilate capillaries by actively relaxing pericytes. Dilation is mediated by prostaglandin E2, but requires nitric oxide release to suppress vasoconstricting 20-HETE synthesis. In vivo, when sensory input increases blood flow, capillaries dilate before arterioles and are estimated to produce 84% of the blood flow increase. In pathology, ischaemia evokes capillary constriction by pericytes. We show that this is followed by pericyte death in rigor, which may irreversibly constrict capillaries and damage the blood-brain barrier. Thus, pericytes are major regulators of cerebral blood flow and initiators of functional imaging signals. Prevention of pericyte constriction and death may reduce the long-lasting blood flow decrease that damages neurons after stroke.
Collapse
Affiliation(s)
- Catherine N Hall
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| | - Clare Reynell
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| | - Bodil Gesslein
- Department of Neuroscience & Pharmacology and Center for Healthy Aging, and Department of Clinical Neurophysiology, Glostrup Hospital, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Nicola B Hamilton
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| | - Anusha Mishra
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| | - Brad A Sutherland
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Fergus M O'Farrell
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Martin Lauritzen
- Department of Neuroscience & Pharmacology and Center for Healthy Aging, and Department of Clinical Neurophysiology, Glostrup Hospital, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology University College London, Gower St., London, WC1E 6BT, UK
| |
Collapse
|
29
|
Shelat PB, Plant LD, Wang JC, Lee E, Marks JD. The membrane-active tri-block copolymer pluronic F-68 profoundly rescues rat hippocampal neurons from oxygen-glucose deprivation-induced death through early inhibition of apoptosis. J Neurosci 2013; 33:12287-99. [PMID: 23884935 PMCID: PMC3721839 DOI: 10.1523/jneurosci.5731-12.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 06/11/2013] [Accepted: 06/14/2013] [Indexed: 01/01/2023] Open
Abstract
Pluronic F-68, an 80% hydrophilic member of the Pluronic family of polyethylene-polypropylene-polyethylene tri-block copolymers, protects non-neuronal cells from traumatic injuries and rescues hippocampal neurons from excitotoxic and oxidative insults. F-68 interacts directly with lipid membranes and restores membrane function after direct membrane damage. Here, we demonstrate the efficacy of Pluronic F-68 in rescuing rat hippocampal neurons from apoptosis after oxygen-glucose deprivation (OGD). OGD progressively decreased neuronal survival over 48 h in a severity-dependent manner, the majority of cell death occurring after 12 h after OGD. Administration of F-68 for 48 h after OGD rescued neurons from death in a dose-dependent manner. At its optimal concentration (30 μm), F-68 rescued all neurons that would have died after the first hour after OGD. This level of rescue persisted when F-68 administration was delayed 12 h after OGD. F-68 did not alter electrophysiological parameters controlling excitability, NMDA receptor-activated currents, or NMDA-induced increases in cytosolic calcium concentrations. However, F-68 treatment prevented phosphatidylserine externalization, caspase activation, loss of mitochondrial membrane potential, and BAX translocation to mitochondria, indicating that F-68 alters apoptotic mechanisms early in the intrinsic pathway of apoptosis. The profound neuronal rescue provided by F-68 after OGD and the high level of efficacy with delayed administration indicate that Pluronic copolymers may provide a novel, membrane-targeted approach to rescuing neurons after brain ischemia. The ability of membrane-active agents to block apoptosis suggests that membranes or their lipid components play prominent roles in injury-induced apoptosis.
Collapse
Affiliation(s)
- Phullara B. Shelat
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Leigh D. Plant
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Janice C. Wang
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Elizabeth Lee
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Jeremy D. Marks
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
30
|
Kohonen P, Benfenati E, Bower D, Ceder R, Crump M, Cross K, Grafström RC, Healy L, Helma C, Jeliazkova N, Jeliazkov V, Maggioni S, Miller S, Myatt G, Rautenberg M, Stacey G, Willighagen E, Wiseman J, Hardy B. The ToxBank Data Warehouse: Supporting the Replacement of In Vivo Repeated Dose Systemic Toxicity Testing. Mol Inform 2013; 32:47-63. [PMID: 27481023 DOI: 10.1002/minf.201200114] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/27/2012] [Indexed: 12/12/2022]
Abstract
The aim of the SEURAT-1 (Safety Evaluation Ultimately Replacing Animal Testing-1) research cluster, comprised of seven EU FP7 Health projects co-financed by Cosmetics Europe, is to generate a proof-of-concept to show how the latest technologies, systems toxicology and toxicogenomics can be combined to deliver a test replacement for repeated dose systemic toxicity testing on animals. The SEURAT-1 strategy is to adopt a mode-of-action framework to describe repeated dose toxicity, combining in vitro and in silico methods to derive predictions of in vivo toxicity responses. ToxBank is the cross-cluster infrastructure project whose activities include the development of a data warehouse to provide a web-accessible shared repository of research data and protocols, a physical compounds repository, reference or "gold compounds" for use across the cluster (available via wiki.toxbank.net), and a reference resource for biomaterials. Core technologies used in the data warehouse include the ISA-Tab universal data exchange format, REpresentational State Transfer (REST) web services, the W3C Resource Description Framework (RDF) and the OpenTox standards. We describe the design of the data warehouse based on cluster requirements, the implementation based on open standards, and finally the underlying concepts and initial results of a data analysis utilizing public data related to the gold compounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lyn Healy
- National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | | | | - Silvia Maggioni
- Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | - Glyn Stacey
- National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | | | |
Collapse
|
31
|
Abstract
Neuronal computation is energetically expensive. Consequently, the brain's limited energy supply imposes constraints on its information processing capability. Most brain energy is used on synaptic transmission, making it important to understand how energy is provided to and used by synapses. We describe how information transmission through presynaptic terminals and postsynaptic spines is related to their energy consumption, assess which mechanisms normally ensure an adequate supply of ATP to these structures, consider the influence of synaptic plasticity and changing brain state on synaptic energy use, and explain how disruption of the energy supply to synapses leads to neuropathology.
Collapse
Affiliation(s)
- Julia J Harris
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | |
Collapse
|
32
|
Anoxic depolarization of hippocampal astrocytes: possible modulation by P2X7 receptors. Neurochem Int 2012; 62:15-22. [PMID: 23147683 DOI: 10.1016/j.neuint.2012.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 12/30/2022]
Abstract
Current responses from CA1 neurons and stratum oriens astrocytes were recorded from hippocampal brain slices by means of the whole-cell patch-clamp technique. Anoxic depolarization (AD) was induced by an oxygen/glucose-deprived (OGD) medium also containing sodium iodoacetate and antimycin, in order to block glycolysis and oxidative phosphorylation, respectively. Anoxic depolarization has been reported to be due to the sudden increase of the extracellular K(+) concentration and the accompanying explosive rise in glutamate concentration. We asked ourselves whether the release of ATP activating P2X7 receptors is also involved in the AD. Although, the AD was evoked in absolute synchrony in neurons and astrocytes, and the NMDA receptor antagonistic AP-5 depressed these responses, neither the non-selective P2 receptor antagonist PPADS, nor the highly selective P2X7 receptor antagonist A438079 interfered with the AD or its delay time in neurons/astrocytes after inducing chemical hypoxia. However, A438079, but not PPADS increased in astrocytes the slow inward current observed in a hypoxic medium. It is concluded that ATP co-released with glutamate by hypoxic stimulation has only a minor function in the present brain slice system.
Collapse
|
33
|
Abstract
Potential roles for lactate in the energetics of brain activation have changed radically during the past three decades, shifting from waste product to supplemental fuel and signaling molecule. Current models for lactate transport and metabolism involving cellular responses to excitatory neurotransmission are highly debated, owing, in part, to discordant results obtained in different experimental systems and conditions. Major conclusions drawn from tabular data summarizing results obtained in many laboratories are as follows: Glutamate-stimulated glycolysis is not an inherent property of all astrocyte cultures. Synaptosomes from the adult brain and many preparations of cultured neurons have high capacities to increase glucose transport, glycolysis, and glucose-supported respiration, and pathway rates are stimulated by glutamate and compounds that enhance metabolic demand. Lactate accumulation in activated tissue is a minor fraction of glucose metabolized and does not reflect pathway fluxes. Brain activation in subjects with low plasma lactate causes outward, brain-to-blood lactate gradients, and lactate is quickly released in substantial amounts. Lactate utilization by the adult brain increases during lactate infusions and strenuous exercise that markedly increase blood lactate levels. Lactate can be an 'opportunistic', glucose-sparing substrate when present in high amounts, but most evidence supports glucose as the major fuel for normal, activated brain.
Collapse
|
34
|
Pellerin L, Magistretti PJ. Sweet sixteen for ANLS. J Cereb Blood Flow Metab 2012; 32:1152-66. [PMID: 22027938 PMCID: PMC3390819 DOI: 10.1038/jcbfm.2011.149] [Citation(s) in RCA: 523] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Revised: 08/24/2011] [Accepted: 09/22/2011] [Indexed: 02/03/2023]
Abstract
Since its introduction 16 years ago, the astrocyte-neuron lactate shuttle (ANLS) model has profoundly modified our understanding of neuroenergetics by bringing a cellular and molecular resolution. Praised or disputed, the concept has never ceased to attract attention, leading to critical advances and unexpected insights. Here, we summarize recent experimental evidence further supporting the main tenets of the model. Thus, evidence for distinct metabolic phenotypes between neurons (mainly oxidative) and astrocytes (mainly glycolytic) have been provided by genomics and classical metabolic approaches. Moreover, it has become clear that astrocytes act as a syncytium to distribute energy substrates such as lactate to active neurones. Glycogen, the main energy reserve located in astrocytes, is used as a lactate source to sustain glutamatergic neurotransmission and synaptic plasticity. Lactate is also emerging as a neuroprotective agent as well as a key signal to regulate blood flow. Characterization of monocarboxylate transporter regulation indicates a possible involvement in synaptic plasticity and memory. Finally, several modeling studies captured the implications of such findings for many brain functions. The ANLS model now represents a useful, experimentally based framework to better understand the coupling between neuronal activity and energetics as it relates to neuronal plasticity, neurodegeneration, and functional brain imaging.
Collapse
Affiliation(s)
- Luc Pellerin
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Pierre J Magistretti
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, and Center for Psychiatric Neuroscience UNIL-CHUV, Lausanne, Switzerland
| |
Collapse
|
35
|
Xiang K, Earl D, Dwyer T, Behrle BL, Tietz EI, Greenfield LJ. Hypoxia enhances high-voltage-activated calcium currents in rat primary cortical neurons via calcineurin. Epilepsy Res 2012; 99:293-305. [PMID: 22245138 PMCID: PMC3341530 DOI: 10.1016/j.eplepsyres.2011.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 12/04/2011] [Accepted: 12/18/2011] [Indexed: 01/09/2023]
Abstract
Hypoxia regulates neuronal ion channels, sometimes resulting in seizures. We evaluated the effects of brief sustained hypoxia (1% O(2), 4h) on voltage-gated calcium channels (VGCCs) in cultured rat primary cortical neurons. High-voltage activated (HVA) Ca(2+) currents were acquired immediately after hypoxic exposure or after 48h recovery in 95% air/5% CO(2). Maximal Ca(2+) current density increased 1.5-fold immediately after hypoxia, but reverted to baseline after 48h normoxia. This enhancement was primarily due to an increase in L-type VGCC activity, since nimodipine-insensitive residual Ca(2+) currents were unchanged. The half-maximal potentials of activation and steady-state inactivation were unchanged. The calcineurin inhibitors FK-506 (in the recording pipette) or cyclosporine A (during hypoxia) prevented the post-hypoxic increase in HVA Ca(2+) currents, while rapamycin and okadaic acid did not. L-type VGCCs were the source of Ca(2+) for calcineurin activation, as nimodipine during hypoxia prevented post-hypoxic enhancement. Hypoxia transiently potentiated L-type VGCC currents via calcineurin, suggesting a positive feedback loop to amplify neuronal calcium signaling that may contribute to seizure generation.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Neurology, University of Toledo College of Medicine, Toledo, OH, USA
| | | | | | | | | | | |
Collapse
|
36
|
Contribution of astrocyte glycogen stores to progression of spreading depression and related events in hippocampal slices. Neuroscience 2011; 192:295-303. [PMID: 21600270 DOI: 10.1016/j.neuroscience.2011.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 05/01/2011] [Accepted: 05/02/2011] [Indexed: 11/23/2022]
Abstract
Spreading depression (SD) is a wave of coordinated cellular depolarization that propagates slowly throughout brain tissue. SD has been associated with migraine aura, and related events have been implicated in the enlargement of some brain injuries. Selective disruption of astrocyte oxidative metabolism has previously been shown to increase the propagation rate of SD in vivo, but it is currently unknown whether astrocyte glycogen stores make significant contributions to the onset or propagation of SD. We examined SD in acutely-prepared murine hippocampal slices, using either localized microinjections of KCl or oxygen and glucose deprivation (OGD) as stimuli. A combination of glycogenolysis inhibitors 1,4-dideoxy-1,4-imino-d-arabinitol (DAB) and 1-deoxynojirimycin (DNJ) increased the propagation rates of both high K(+)-SD and OGD-SD. Consistent with these observations, exposure to l-methionine-dl-sulfoximine (MSO) increased slice glycogen levels and decreased OGD-SD propagation rates. Effects of glycogen depletion were matched by selective inhibition of astrocyte tricarboxylic acid (TCA) cycle activity by fluoroacetate (FA). Prolonged exposure to reduced extracellular glucose (2 mM) has been suggested to deplete slice glycogen stores, but significant modification SD of propagation rate was not observed with this treatment. Furthermore, decreases in OGD-SD latency with this preexposure paradigm appeared to be due to depletion of glucose, rather than glycogen availability. These results suggest that astrocyte glycogen stores contribute to delaying the advancing wavefront of SD, including during the severe metabolic challenge of OGD. Approaches to enhance astrocyte glycogen reserves could be beneficial for delaying or preventing SD in some pathologic conditions.
Collapse
|
37
|
Zhang H, Cao HJ, Kimelberg HK, Zhou M. Volume regulated anion channel currents of rat hippocampal neurons and their contribution to oxygen-and-glucose deprivation induced neuronal death. PLoS One 2011; 6:e16803. [PMID: 21347298 PMCID: PMC3037944 DOI: 10.1371/journal.pone.0016803] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 12/30/2010] [Indexed: 01/29/2023] Open
Abstract
Volume-regulated anion channels (VRAC) are widely expressed chloride channels that are critical for the cell volume regulation. In the mammalian central nervous system, the physiological expression of neuronal VRAC and its role in cerebral ischemia are issues largely unknown. We show that hypoosmotic medium induce an outwardly rectifying chloride conductance in CA1 pyramidal neurons in rat hippocampal slices. The induced chloride conductance was sensitive to some of the VRAC inhibitors, namely, IAA-94 (300 µM) and NPPB (100 µM), but not to tamoxifen (10 µM). Using oxygen-and-glucose deprivation (OGD) to simulate ischemic conditions in slices, VRAC activation appeared after OGD induced anoxic depolarization (AD) that showed a progressive increase in current amplitude over the period of post-OGD reperfusion. The OGD induced VRAC currents were significantly inhibited by inhibitors for glutamate AMPA (30 µM NBQX) and NMDA (40 µM AP-5) receptors in the OGD solution, supporting the view that induction of AD requires an excessive Na+-loading via these receptors that in turn to activate neuronal VRAC. In the presence of NPPB and DCPIB in the post-OGD reperfusion solution, the OGD induced CA1 pyramidal neuron death, as measured by TO-PRO-3-I staining, was significantly reduced, although DCPIB did not appear to be an effective neuronal VRAC blocker. Altogether, we show that rat hippocampal pyramidal neurons express functional VRAC, and ischemic conditions can initial neuronal VRAC activation that may contribute to ischemic neuronal damage.
Collapse
Affiliation(s)
- Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People‚s Republic of China
- * E-mail: (HZ); (MZ)
| | - H. James Cao
- Ordway Research Institute, Albany, New York, United States of America
| | | | - Min Zhou
- Ordway Research Institute, Albany, New York, United States of America
- * E-mail: (HZ); (MZ)
| |
Collapse
|
38
|
Madry C, Haglerød C, Attwell D. The role of pannexin hemichannels in the anoxic depolarization of hippocampal pyramidal cells. ACTA ACUST UNITED AC 2010; 133:3755-63. [PMID: 20940167 PMCID: PMC2995884 DOI: 10.1093/brain/awq284] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Neuronal gap junctional hemichannels, composed of pannexin-1 subunits, have been suggested to play a crucial role in epilepsy and brain ischaemia. After a few minutes of anoxia or ischaemia, neurons in brain slices show a rapid depolarization to ∼−20 mV, called the anoxic depolarization. Glutamate receptor blockers can prevent the anoxic depolarization, suggesting that it is produced by a cation influx through glutamate-gated channels. However, in isolated hippocampal pyramidal cells, simulated ischaemia evokes a large inward current and an increase in permeability to large molecules, mediated by the opening of pannexin-1 hemichannels. N-methyl-d-aspartate is also reported to open these hemichannels, suggesting that the activation of N-methyl-d-aspartate receptors, which occurs when glutamate is released in ischaemia, might cause the anoxic depolarization by evoking a secondary ion flux through pannexin-1 hemichannels. We tested the contribution of pannexin hemichannels to the anoxic depolarization in CA1 pyramidal cells in the more physiological environment of hippocampal slices. Three independent inhibitors of hemichannels—carbenoxolone, lanthanum and mefloquine—had no significant effect on the current generating the anoxic depolarization, while a cocktail of glutamate and gamma-aminobutyric acid class A receptor blockers abolished it. We conclude that pannexin hemichannels do not generate the large inward current that underlies the anoxic depolarization. Glutamate receptor channels remain the main candidate for generating the large inward current that produces the anoxic depolarization.
Collapse
Affiliation(s)
- Christian Madry
- Department of Physiology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | | |
Collapse
|
39
|
Yu AS, Hirayama BA, Timbol G, Liu J, Basarah E, Kepe V, Satyamurthy N, Huang SC, Wright EM, Barrio JR. Functional expression of SGLTs in rat brain. Am J Physiol Cell Physiol 2010; 299:C1277-84. [PMID: 20826762 DOI: 10.1152/ajpcell.00296.2010] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This work provides evidence of previously unrecognized uptake of glucose via sodium-coupled glucose transporters (SGLTs) in specific regions of the brain. The current understanding of functional glucose utilization in brain is largely based on studies using positron emission tomography (PET) with the glucose tracer 2-deoxy-2-[F-18]fluoro-D-glucose (2-FDG). However, 2-FDG is only a good substrate for facilitated-glucose transporters (GLUTs), not for SGLTs. Thus, glucose accumulation measured by 2-FDG omits the role of SGLTs. We designed and synthesized two high-affinity tracers: one, α-methyl-4-[F-18]fluoro-4-deoxy-D-glucopyranoside (Me-4FDG), is a highly specific SGLT substrate and not transported by GLUTs; the other one, 4-[F-18]fluoro-4-deoxy-D-glucose (4-FDG), is transported by both SGLTs and GLUTs and will pass through the blood brain barrier (BBB). In vitro Me-4FDG autoradiography was used to map the distribution of uptake by functional SGLTs in brain slices with a comparable result from in vitro 4-FDG autoradiography. Immunohistochemical assays showed that uptake was consistent with the distribution of SGLT protein. Ex vivo 4-FDG autoradiography showed that SGLTs in these areas are functionally active in the normal in vivo brain. The results establish that SGLTs are a normal part of the physiology of specific areas of the brain, including hippocampus, amygdala, hypothalamus, and cerebral cortices. 4-FDG PET imaging also established that this BBB-permeable SGLT tracer now offers a functional imaging approach in humans to assess regulation of SGLT activity in health and disease.
Collapse
Affiliation(s)
- Amy S Yu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, Universityof California Los Angeles, California 90095-1751, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Denes A, Thornton P, Rothwell NJ, Allan SM. Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain Behav Immun 2010; 24:708-23. [PMID: 19770034 DOI: 10.1016/j.bbi.2009.09.010] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a classical host defence response to infection and injury that has many beneficial effects. However, inappropriate (in time, place and magnitude) inflammation is increasingly implicated in diverse disease states, now including cancer, diabetes, obesity, atherosclerosis, heart disease and, most relevant here, CNS disease. A growing literature shows strong correlations between inflammatory status and the risk of cerebral ischaemia (CI, most commonly stroke), as well as with outcome from an ischaemic event. Intervention studies to demonstrate a causal link between inflammation and CI (or its consequences) are limited but are beginning to emerge, while experimental studies of CI have provided direct evidence that key inflammatory mediators (cytokines, chemokines and inflammatory cells) contribute directly to ischaemic brain injury. However, it remains to be determined what the relative importance of systemic (largely peripheral) versus CNS inflammation is in CI. Animal models in which CI is driven by a CNS intervention may not accurately reflect the clinical condition; stroke being typically induced by atherosclerosis or cardiac dysfunction, and hence current experimental paradigms may underestimate the contribution of peripheral inflammation. Experimental studies have already identified a number of potential anti-inflammatory therapeutic interventions that may limit ischaemic brain damage, some of which have been tested in early clinical trials with potentially promising results. However, a greater understanding of the contribution of inflammation to CI is still required, and this review highlights some of the key mechanism that may offer future therapeutic targets.
Collapse
Affiliation(s)
- A Denes
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
41
|
Abstract
In addition to their role in providing myelin for rapid impulse propagation, the glia that ensheath long axons are required for the maintenance of normal axon transport and long-term survival. This presumably ancestral function seems to be independent of myelin membrane wrapping. Here, I propose that ensheathing glia provide trophic support to axons that are metabolically isolated, and that myelin itself might cause such isolation. This glial support of axonal integrity may be relevant for a number of neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Klaus-Armin Nave is at the Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Herrmann-Rein-Strasse 3, D-37075 Goettingen, Germany.
| |
Collapse
|
42
|
HPLC-UV measurements of metabolites in the supernatant of endothelial cells exposed to oxidative stress. Anal Bioanal Chem 2010; 396:1763-71. [DOI: 10.1007/s00216-009-3398-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 12/10/2009] [Indexed: 01/08/2023]
|
43
|
Izumi Y, Zorumski CF. Glial-neuronal interactions underlying fructose utilization in rat hippocampal slices. Neuroscience 2009; 161:847-54. [PMID: 19362122 DOI: 10.1016/j.neuroscience.2009.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 03/18/2009] [Accepted: 04/04/2009] [Indexed: 12/21/2022]
Abstract
Although fructose is commonly used as a sweetener, its effects on brain function are unclear. Using rat hippocampal slices, we found that fructose and mannose, like pyruvate, preserve ATP levels during 3-h of glucose deprivation. Similarly, fructose and mannose restored synaptic potentials (excitatory postsynaptic potential, EPSPs) depressed during glucose deprivation. However, restoration of synaptic responses was slow and only partial with fructose. EPSPs supported by mannose were inhibited by cytochalasin B (CCB), a glucose transport inhibitor, but were not inhibited by alpha-cyano-4-hydroxycinnamate (4-CIN), a monocarboxylate transport inhibitor, indicating that neurons use mannose via glucose transporters. In contrast, both CCB and 4-CIN depressed EPSPs supported by fructose, suggesting that fructose may be taken up by non-neuronal cells through CCB sensitive hexose transporters and metabolized to a monocarboxylate for subsequent use during neuronal respiration. Supporting this possibility, 20 minutes of oxygen deprivation in the presence of fructose resulted in functional and morphological deterioration whereas oxygen deprivation in the presence of glucose or mannose had minimal toxic effects. These results indicate that neuronal fructose utilization differs from glucose and mannose and likely involves release of monocarboxylates from glia.
Collapse
Affiliation(s)
- Y Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | |
Collapse
|
44
|
Morgenthaler FD, Lanz BR, Petit JM, Frenkel H, Magistretti PJ, Gruetter R. Alteration of brain glycogen turnover in the conscious rat after 5h of prolonged wakefulness. Neurochem Int 2009; 55:45-51. [PMID: 19428806 DOI: 10.1016/j.neuint.2009.02.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 02/23/2009] [Accepted: 02/27/2009] [Indexed: 02/04/2023]
Abstract
Although glycogen (Glyc) is the main carbohydrate storage component, the role of Glyc in the brain during prolonged wakefulness is not clear. The aim of this study was to determine brain Glyc concentration ([]) and turnover time (tau) in euglycemic conscious and undisturbed rats, compared to rats maintained awake for 5h. To measure the metabolism of [1-(13)C]-labeled Glc into Glyc, 23 rats received a [1-(13)C]-labeled Glc solution as drink (10% weight per volume in tap water) ad libitum as their sole source of exogenous carbon for a "labeling period" of either 5h (n=13), 24h (n=5) or 48 h (n=5). Six of the rats labeled for 5h were continuously maintained awake by acoustic, tactile and olfactory stimuli during the labeling period, which resulted in slightly elevated corticosterone levels. Brain [Glyc] measured biochemically after focused microwave fixation in the rats maintained awake (3.9+/-0.2 micromol/g, n=6) was not significantly different from that of the control group (4.0+/-0.1 micromol/g, n=7; t-test, P>0.5). To account for potential variations in plasma Glc isotopic enrichment (IE), Glyc IE was normalized by N-acetyl-aspartate (NAA) IE. A simple mathematical model was developed to derive brain Glyc turnover time as 5.3h with a fit error of 3.2h and NAA turnover time as 15.6h with a fit error of 6.5h, in the control rats. A faster tau(Glyc) (2.9h with a fit error of 1.2h) was estimated in the rats maintained awake for 5h. In conclusion, 5h of prolonged wakefulness mainly activates glycogen metabolism, but has minimal effect on brain [Glyc].
Collapse
Affiliation(s)
- Florence D Morgenthaler
- Centre d'Imagerie Biomédicale, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | |
Collapse
|
45
|
Morgenthaler FD, van Heeswijk RB, Xin L, Laus S, Frenkel H, Lei H, Gruetter R. Non-invasive quantification of brain glycogen absolute concentration. J Neurochem 2008; 107:1414-23. [PMID: 19013831 DOI: 10.1111/j.1471-4159.2008.05717.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The only currently available method to measure brain glycogen in vivo is 13C NMR spectroscopy. Incorporation of 13C-labeled glucose (Glc) is necessary to allow glycogen measurement, but might be affected by turnover changes. Our aim was to measure glycogen absolute concentration in the rat brain by eliminating label turnover as variable. The approach is based on establishing an increased, constant 13C isotopic enrichment (IE). 13C-Glc infusion is then performed at the IE of brain glycogen. As glycogen IE cannot be assessed in vivo, we validated that it can be inferred from that of N-acetyl-aspartate IE in vivo: After [1-13C]-Glc ingestion, glycogen IE was 2.2 +/- 0.1 fold that of N-acetyl-aspartate (n = 11, R(2) = 0.77). After subsequent Glc infusion, glycogen IE equaled brain Glc IE (n = 6, paired t-test, p = 0.37), implying isotopic steady-state achievement and complete turnover of the glycogen molecule. Glycogen concentration measured in vivo by 13C NMR (mean +/- SD: 5.8 +/- 0.7 micromol/g) was in excellent agreement with that in vitro (6.4 +/- 0.6 micromol/g, n = 5). When insulin was administered, the stability of glycogen concentration was analogous to previous biochemical measurements implying that glycogen turnover is activated by insulin. We conclude that the entire glycogen molecule is turned over and that insulin activates glycogen turnover.
Collapse
Affiliation(s)
- Florence D Morgenthaler
- Centre d'Imagerie Biomédicale (CIBM), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
46
|
Chen PH, Cai WQ, Wang LY, Deng QY. A morphological and electrophysiological study on the postnatal development of oligodendrocyte precursor cells in the rat brain. Brain Res 2008; 1243:27-37. [DOI: 10.1016/j.brainres.2008.09.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Revised: 08/18/2008] [Accepted: 09/10/2008] [Indexed: 12/29/2022]
|
47
|
Zhang H, Xie M, Schools GP, Feustel PF, Wang W, Lei T, Kimelberg HK, Zhou M. Tamoxifen mediated estrogen receptor activation protects against early impairment of hippocampal neuron excitability in an oxygen/glucose deprivation brain slice ischemia model. Brain Res 2008; 1247:196-211. [PMID: 18992727 DOI: 10.1016/j.brainres.2008.10.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 10/02/2008] [Accepted: 10/04/2008] [Indexed: 12/18/2022]
Abstract
Pretreatment of ovarectomized rats with estrogen shows long-term protection via activation of the estrogen receptor (ER). However, it remains unknown whether activation of the ER can provide protection against early neuronal damage when given acutely. We simulated ischemic conditions by applying oxygen and glucose deprived (OGD) solution to acute male rat hippocampal slices and examined the neuronal electrophysiological changes. Pyramidal neurons and interneurons showed a time-dependent membrane potential depolarization and reduction in evoked action potential frequency and amplitude over a 10 to 15 min OGD exposure. These changes were largely suppressed by 10 microM TAM. The TAM effect was neuron-specific as the OGD-induced astrocytic membrane potential depolarization was not altered. The TAM effect was mediated through ER activation because it could be simulated by 17beta-estradiol and was completely inhibited by the ER inhibitor ICI 182, 780, and is therefore an example of TAM's selective estrogen receptor modulator (SERM) action. We further show that TAM's effects on OGD-induced impairment of neuronal excitability was largely due to activation of neuroprotective BK channels, as the TAM effect was markedly attenuated by the BK channel inhibitor paxilline at 10 microM. TAM also significantly reduced the frequency and amplitude of AMPA receptor mediated spontaneous excitatory postsynaptic currents (sEPSCs) in pyramidal neurons which is an early consequence of OGD. Altogether, this study demonstrates that both 17beta-estradiol and TAM attenuate neuronal excitability impairment early on in a simulated ischemia model via ER activation mediated potentiation of BK K(+) channels and reduction in enhanced neuronal AMPA/NMDA receptor-mediated excitotoxicity.
Collapse
Affiliation(s)
- Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, 430030, PR China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhang L, Deng T, Sun Y, Liu K, Yang Y, Zheng X. Role for nitric oxide in permeability of hippocampal neuronal hemichannels during oxygen glucose deprivation. J Neurosci Res 2008; 86:2281-91. [PMID: 18381763 DOI: 10.1002/jnr.21675] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Increased hemichannel opening induced by oxygen glucose deprivation (OGD) was reported in the hippocampal pyramidal neuron. It was suggested that the pannexin1 hemichannel opening could mediate ionic flux dysregulation, anoxic depolarization, and energy-depleting efflux of glucose and ATP for ischemic neurons. However, the regulatory mechanisms of pannexin1 hemichannel opening have been poorly understood. Here we showed that excessive generation of nitric oxide (NO) during ischemia could induce the calcein leakage from neurons, which was markedly reduced by NO synthase inhibitor. The calcein leakage from neurons during OGD was also attenuated by the application of N-ethylmaleimide (NEM), an SH-alkylating agent, and dithiothreitol (DTT), a reducer of oxidized sulfhydryl groups. However, the soluble guanylyl cyclase (sGC) inhibitor had a minor effect on the calcein leakage during OGD. Furthermore, the elevated intracellular but not extracellular levels of glutathione could also inhibit the calcein leakage during OGD. Similar results were observed in metabolic inhibition (MI), which is another ischemic-like condition. Finally, immunocytochemical and immunoblotting analysis revealed that, after 1 hr of OGD stimulation, the distribution and expression of pannexin1 showed no significant difference compared with control. However, the pannexin1 mRNA expression was elevated after 1 hr of OGD and a sustained increase was maintained during reperfusion. These results implied that the reactive oxygen species (ROS), especially NO, might be involved in the enhanced pannexin1 hemichannel opening and that the S-nitrosylation but not the NO/cGMP pathway played a more important role in this event.
Collapse
Affiliation(s)
- Le Zhang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | |
Collapse
|
49
|
Furness DN, Dehnes Y, Akhtar AQ, Rossi DJ, Hamann M, Grutle NJ, Gundersen V, Holmseth S, Lehre KP, Ullensvang K, Wojewodzic M, Zhou Y, Attwell D, Danbolt NC. A quantitative assessment of glutamate uptake into hippocampal synaptic terminals and astrocytes: new insights into a neuronal role for excitatory amino acid transporter 2 (EAAT2). Neuroscience 2008; 157:80-94. [PMID: 18805467 DOI: 10.1016/j.neuroscience.2008.08.043] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2007] [Revised: 08/07/2008] [Accepted: 08/08/2008] [Indexed: 01/04/2023]
Abstract
The relative distribution of the excitatory amino acid transporter 2 (EAAT2) between synaptic terminals and astroglia, and the importance of EAAT2 for the uptake into terminals is still unresolved. Here we have used antibodies to glutaraldehyde-fixed d-aspartate to identify electron microscopically the sites of d-aspartate accumulation in hippocampal slices. About 3/4 of all terminals in the stratum radiatum CA1 accumulated d-aspartate-immunoreactivity by an active dihydrokainate-sensitive mechanism which was absent in EAAT2 glutamate transporter knockout mice. These terminals were responsible for more than half of all d-aspartate uptake of external substrate in the slices. This is unexpected as EAAT2-immunoreactivity observed in intact brain tissue is mainly associated with astroglia. However, when examining synaptosomes and slice preparations where the extracellular space is larger than in perfusion fixed tissue, it was confirmed that most EAAT2 is in astroglia (about 80%). Neither d-aspartate uptake nor EAAT2 protein was detected in dendritic spines. About 6% of the EAAT2-immunoreactivity was detected in the plasma membrane of synaptic terminals (both within and outside of the synaptic cleft). Most of the remaining immunoreactivity (8%) was found in axons where it was distributed in a plasma membrane surface area several times larger than that of astroglia. This explains why the densities of neuronal EAAT2 are low despite high levels of mRNA in CA3 pyramidal cell bodies, but not why EAAT2 in terminals account for more than half of the uptake of exogenous substrate by hippocampal slice preparations. This and the relative amount of terminal versus glial uptake in the intact brain remain to be discovered.
Collapse
Affiliation(s)
- D N Furness
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffs, ST5 5BG, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kawauchi S, Sato S, Ooigawa H, Nawashiro H, Ishihara M, Kikuchi M. Simultaneous measurement of changes in light absorption due to the reduction of cytochrome c oxidase and light scattering in rat brains during loss of tissue viability. APPLIED OPTICS 2008; 47:4164-76. [PMID: 18670575 DOI: 10.1364/ao.47.004164] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
We performed the simultaneous measurement of intrinsic optical signals (IOSs) related to metabolic activity and cellular and subcellular morphological characteristics, i.e., light scattering for a rat global ischemic brain model made by rapidly removing blood by saline infusion. The signals were measured on the basis of multiwavelength diffuse reflectances in which 605 and 830 nm were used to detect the IOSs that are thought to be dominantly affected by redox changes of heme aa(3) and CuA in cytochrome c oxidase (CcO), respectively. For measuring the scattering signal, the wavelength that was found to be most insensitive to the absorption changes, e.g., approximately 620 nm, was used. The measurements suggested that an increase in the absorption due to reduction of heme aa(3) occurred soon after blood clearance, and this was followed by a large triphasic change in light scattering, during which time a decrease in the absorption due to reduction of CuA occurred. Through the triphasic scattering change, scattering signals increased by 5.2 +/- 1.5% (n = 5), and the increase in light scattering showed significant correlation with both the reflectance intensity changes at 605 and 830 nm. This suggests that morphological changes in cells correlate with reductions of heme aa(3) and CuA. Histological analysis of tissue after the triphasic scattering change showed no alteration in either the nuclei or the cytoskeleton, but electron microscopic observation revealed deformed, enlarged mitochondria and expanded dendrites. These findings suggest that the simultaneous measurement of absorption signals related to the redox changes in the CcO and the scattering signal is useful for monitoring tissue viability in the brain.
Collapse
Affiliation(s)
- Satoko Kawauchi
- Department of Medical Engineering, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | | | | | | | | | | |
Collapse
|