Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Thomas J. Sick is active.

Publication


Featured researches published by Thomas J. Sick.


Glia | 2006

Increased seizure duration and slowed potassium kinetics in mice lacking aquaporin-4 water channels

Devin K. Binder; Xiaoming Yao; Zsolt Zador; Thomas J. Sick; A. S. Verkman; Geoffrey T. Manley

The glial water channel aquaporin‐4 (AQP4) has been hypothesized to modulate water and potassium fluxes associated with neuronal activity. In this study, we examined the seizure phenotype of AQP4 −/− mice using in vivo electrical stimulation and electroencephalographic (EEG) recording. AQP4 −/− mice were found to have dramatically prolonged stimulation‐evoked seizures after hippocampal stimulation compared to wild‐type controls (33 ± 2 s vs. 13 ± 2 s). In addition, AQP4 −/− mice were found to have a higher seizure threshold (167 ± 17 μA vs. 114 ± 10 μA). To assess a potential effect of AQP4 on potassium kinetics, we used in vivo recording with potassium‐sensitive microelectrodes after direct cortical stimulation. Although there was no significant difference in baseline or peak [K+]o, the rise time to peak [K+]o (t1/2, 2.3 ± 0.5 s) as well as the recovery to baseline [K+]o (t1/2, 15.6 ± 1.5 s) were slowed in AQP4 −/− mice compared to WT mice (t1/2, 0.5 ± 0.1 and 6.6 ± 0.7 s, respectively). These results implicate AQP4 in the expression and termination of seizure activity and support the hypothesis that AQP4 is coupled to potassium homeostasis in vivo.


Journal of Cerebral Blood Flow and Metabolism | 1997

Rapid preconditioning protects rats against ischemic neuronal damage after 3 but not 7 days of reperfusion following global cerebral ischemia

Miguel A. Perez-Pinzon; Guang Ping Xu; W. Dalton Dietrich; Myron Rosenthal; Thomas J. Sick

Earlier studies indicated that sublethal ischemic insults separated by many hours may “precondition” and, thereby, protect tissues from subsequent insults. In Wistar rats, we examined the hypothesis that ischemic preconditioning (IPC) can improve histopathological outcome even if the “conditioning” and “test” ischemic insults are separated by only 30 min. Normothermic (36.5–37°C) global cerebral ischemia was produced by bilateral carotid artery ligation after lowering mean systemic blood pressure. The conditioning ischemic insult lasted 2 min and was associated with a time sufficient to provoke “anoxic depolarization” (AD) (i.e., the abrupt maximal increase in extracellular potassium ion activity). After 30 min of reperfusion, 10-min test ischemia was produced, and histopathology was assessed 3 and 7 days later. After 3 days of reperfusion, neuroprotection was most robust in the left lateral, middle and medial subsections of the hippocampal CA1 subfield and in the cortex, where protection was 91, 76, 70 and 86%, respectively. IPC also protected the right lateral, middle and medial subsections of the hippocampal CA1 region. These data demonstrate that neuroprotection against acute neuronal injury can be achieved by conditioning insults followed by only short (30 min) periods of reperfusion. However, neuroprotection almost disappeared when reperfusion was continued for 7 days. When test ischemia was decreased to 7 min, a clear trend of neuroprotection by IPC was observed. These data suggest that subsequent rescue of neuronal populations could be achieved with better understanding of the neuroprotective mechanisms involved in this rapid IPC model.


Journal of Cerebral Blood Flow and Metabolism | 2001

Protein Aggregation after Focal Brain Ischemia and Reperfusion

Bing Ren Hu; Shorena Janelidze; Myron D. Ginsberg; Raul Busto; Miguel A. Perez-Pinzon; Thomas J. Sick; Bo K. Siesjö; Chunli Liu

Two hours of transient focal brain ischemia causes acute neuronal death in the striatal core region and a somewhat more delayed type of neuronal death in neocortex. The objective of the current study was to investigate protein aggregation and neuronal death after focal brain ischemia in rats. Brain ischemia was induced by 2 hours of middle cerebral artery occlusion. Protein aggregation was analyzed by electron microscopy, laser-scanning confocal microscopy, and Western blotting. Two hours of focal brain ischemia induced protein aggregation in ischemic neocortical neurons at 1 hour of reperfusion, and protein aggregation persisted until neuronal death at 24 hours of reperfusion. Protein aggregates were found in the neuronal soma, dendrites, and axons, and they were associated with intracellular membranous structures during the postischemic phase. High-resolution confocal microscopy showed that clumped protein aggregates surrounding nuclei and along dendrites were formed after brain ischemia. On Western blots, ubiquitinated proteins (ubi-proteins) were dramatically increased in neocortical tissues in the postischemic phase. The ubi-proteins were Triton-insoluble, indicating that they might be irreversibly aggregated. The formation of ubi-protein aggregates after ischemia correlated well with the observed decrease in free ubiquitin and neuronal death. The authors concluded that proteins are severely damaged and aggregated in neurons after focal ischemia. The authors propose that protein damage or aggregation may contribute to ischemic neuronal death.


Journal of Cerebral Blood Flow and Metabolism | 1999

Cytochrome C Is Released From Mitochondria Into the Cytosol After Cerebral Anoxia or Ischemia

Miguel A. Perez-Pinzon; Guang Ping Xu; James G. Born; José Lorenzo; Raul Busto; Myron Rosenthal; Thomas J. Sick

Mitochondrial dysfunction may underlie both acute and delayed neuronal cell death resulting from cerebral ischemia. Specifically, postischemic release of mitochondrial constituents such as the pro-apoptotic respiratory chain component cytochrome c could contribute acutely to further mitochondrial dysfunction and to promote delayed neuronal death. Experiments reported here tested the hypothesis that ischemia or severe hypoxia results in release of cytochrome c from mitochondria. Cytochrome c was measured spectrophotometrically from either the cytosolic fraction of cortical brain homogenates after global ischemia plus reperfusion, or from brain slices subjected to severe hypoxia plus reoxygenation. Cytochrome c content in cytosol derived from cerebral cortex was increased after ischemia and reperfusion. In intact hippocampal slices, there was a loss of reducible cytochrome c after hypoxia/reoxygenation, which is consistent with a decrease of this redox carrier in the mitochondrial pool. These results suggest that cytochrome c is lost to the cytosol after cerebral ischemia in a manner that may contribute to postischemic mitochondrial dysfunction and to delayed neuronal death.


Journal of Cerebral Blood Flow and Metabolism | 2004

Epsilon Protein Kinase C Mediated Ischemic Tolerance Requires Activation of the Extracellular Regulated Kinase Pathway in the Organotypic Hippocampal Slice

Christian Lange-Asschenfeldt; Ami P. Raval; Kunjan R. Dave; Daria Mochly-Rosen; Thomas J. Sick; Miguel A. Perez-Pinzon

Ischemic preconditioning (IPC) promotes brain tolerance against subsequent ischemic insults. Using the organotypic hippocampal slice culture, we conducted the present study to investigate (1) the role of adenosine A1 receptor (A1AR) activation in IPC induction, (2) whether epsilon protein kinase C (ɛPKC) activation after IPC is mediated by the phosphoinositol pathway, and (3) whether ɛPKC protection is mediated by the extracellular signal-regulated kinase (ERK) pathway. Our results demonstrate that activation of A1AR emulated IPC, whereas blockade of the A1AR during IPC diminished neuroprotection. The neuroprotection promoted by the A1AR was also reduced by the ɛPKC antagonist. To determine whether ɛPKC activation in IPC and A1AR preconditioning is mediated by activation of the phosphoinositol pathway, we incubated slices undergoing IPC or adenosine treatment with a phosphoinositol phospholipase C inhibitor. In both cases, preconditioning neuroprotection was significantly attenuated. To further characterize the subsequent signal transduction pathway that ensues after ɛPKC activation, mitogen-activated protein kinase kinase was blocked during IPC and pharmacologic preconditioning (PPC) (with ɛPKC, NMDA, or A1AR agonists). This treatment significantly attenuated IPC- and PPC-induced neuroprotection. In conclusion, we demonstrate that ɛPKC activation after IPC/PPC is essential for neuroprotection against oxygen/glucose deprivation in organotypic slice cultures and that the ERK pathway is downstream to ɛPKC.


Journal of Cerebral Blood Flow and Metabolism | 2001

Ischemic Preconditioning Preserves Mitochondrial Function After Global Cerebral Ischemia in Rat Hippocampus

Kunjan R. Dave; Isabel Saul; Raul Busto; Myron D. Ginsberg; Thomas J. Sick; Miguel A. Perez-Pinzon

Ischemic tolerance in brain develops when sublethal ischemic insults occur before “lethal” cerebral ischemia. Two windows for the induction of tolerance by ischemic preconditioning (IPC) have been proposed: one that occurs within 1 hour after IPC, and another that occurs 1 or 2 days after IPC. The authors tested the hypotheses that IPC would reduce or prevent ischemia-induced mitochondrial dysfunction. IPC and ischemia were produced by bilateral carotid occlusions and systemic hypotension (50 mm Hg) for 2 and 10 minutes, respectively. Nonsynaptosomal mitochondria were harvested 24 hours after the 10-minute “test” ischemic insult. No significant changes were observed in the oxygen consumption rates and activities for hippocampal mitochondrial complexes I to IV between the IPC and sham groups. Twenty-four hours of reperfusion after 10 minutes of global ischemia (without IPC) promoted significant decreases in the oxygen consumption rates in presence of substrates for complexes I and II compared with the IPC and sham groups. These data suggest that IPC protects the integrity of mitochondrial oxidative phosphorylation after cerebral ischemia.


Brain Research | 2000

Chronic failure in the maintenance of long-term potentiation following fluid percussion injury in the rat

Matthew J. Sanders; Thomas J. Sick; Miguel A. Perez-Pinzon; W. Dalton Dietrich; Edward J. Green

Traumatic brain injury (TBI) can produce chronic cognitive learning/memory deficits that are thought to be mediated, in part, by impaired hippocampal function. Experimentally induced TBI is associated with deficits in hippocampal synaptic plasticity (long-term potentiation, or LTP) at acute post-injury intervals but plasticity has not been examined at long-term survival periods. The present study was conducted to assess the temporal profile of LTP after injury and to evaluate the effects of injury severity on plasticity. Separate groups of rats were subjected to mild (1.1-1.4 atm), moderate (1.8-2.1 atm), or severe (2.2-2.7 atm) fluid percussion (FP) injury (or sham surgery) and processed for hippocampal electrophysiology in the first or eighth week after injury. LTP was defined as a lasting increase in field excitatory post-synaptic potential (fEPSP) slope in area CA1 following tetanic stimulation of the Schaffer collaterals. The fEPSP slope was measured for 60 min after tetanus. Assessment of LTP at the acute interval (6 days) revealed modest peak slope potentiation values (129-139%), which declined in each group (including sham) over the hour-long recording session and did not differ between groups. Eight weeks following injury, slices from all groups exhibited robust maximal potentiation (134-147%). Levels of potentiation among groups were similar at the 5-min test interval but differed significantly at the 30- and 60-min test intervals. Whereas sham slices showed stable potentiation for the entire 60-min assessment period, slices in all of the injury groups exhibited a significant decline in potentiation over this period. These experiments reveal a previously unknown effect of TBI whereby experimentally induced injury results in a chronic inability of the CA1 hippocampus to maintain synaptic plasticity. They also provide evidence that sham surgical procedures can significantly influence hippocampal physiology at the acute post-TBI intervals. The results have implications for the mechanisms underlying the impaired synaptic plasticity following TBI.


Brain Research | 1987

Extracellular potassium ion activity and electrophysiology in the hippocampal slice: paradoxical recovery of synaptic transmission during anoxia

Thomas J. Sick; Emily L. Solow; Eugene L. Roberts

The relationship between extracellular potassium ion activity and neuronal excitability during anoxia was investigated in hippocampal slices in vitro. Extracellular field potentials and K+ activity were measured with double-barreled ion-selective microelectrodes placed either in the stratum pyramidale or stratum radiatum of field CA1. Orthodromic spike activity of CA1 pyramidal cells and field excitatory postsynaptic potentials (f-EPSPs) failed rapidly after anoxia with little change in potassium ion activity and without failure of the Schaffer collateral prevolley or antidromic responses of pyramidal cells. As [K+]o approached 8-10 mM, f-EPSPs and orthodromic spike activity recovered spontaneously. Continued anoxia resulted in massive release of K+ into the extracellular space and complete electrical silence. Presynaptic activity and antidromically elicited spike activity recovered promptly upon reoxygenation after anoxia, but synaptic transmission remained blocked for many minutes. Spontaneous recovery of f-EPSPs and spike activity suggests that a simple mechanism involving depolarization or hyperpolarization of neuronal elements cannot account for failure of synaptic transmission observed during anoxia. However, continued elevation of [K+]o and the associated loss of pre- and postsynaptic excitability with more prolonged anoxia indicated that depolarization was responsible for the eventual electrical silence as anoxia progressed.


Brain Research | 2002

Improvement in neuronal survival after ischemic preconditioning in hippocampal slice cultures

Guang Ping Xu; Kunjan R. Dave; Richard J. Vivero; Rainald Schmidt-Kastner; Thomas J. Sick; Miguel A. Perez-Pinzon

The main goals of the current study were to assess: (a) whether a sublethal ischemic insult could protect the CA1 subregion of the hippocampus in organotypic slices against a lethal ischemic insult; and (b) whether this protection is long lasting as determined with an accurate immunohistochemical neuronal marker, NeuN. Hippocampal slice cultures were grown for 12-14 days in vitro. Slices were exposed either to oxygen/glucose deprivation (OGD) for 45 min (ischemia), or OGD for 15 min (ischemic preconditioning), 48 h prior to 45 min OGD, or were untreated (sham). Cell death was estimated by propidium iodide fluorescence 1 day after OGD and by NeuN immunohistochemistry 7 days after OGD. Image analysis was employed to measure the relative optical density of the NeuN-signal in all groups. After ischemia, damaged neurons were shrunken or lost and NeuN immunoreactivity was reduced. Relative optical density of NeuN (ROD [NeuN]) was 0.193+/-0.015 in control (sham) (n=9). In slices that underwent ischemia, ROD [NeuN] declined to 0.108+/-0.018 (n=5) in CA1 (*P<0.05 ROD [NeuN] in preconditioned slice cultures was 0.190+/-0.037 (76% higher than the ischemia group). Similar results were found after measuring PI fluorescence. In the CA1 sub-region, PI fluorescence was about 13, 47 and 17% in the sham, ischemic and IPC groups, respectively. We suggest that the immunohistochemical approach validates the dye uptake method used in slice cultures and yields quantitative data specific for neurons. We also conclude that the organotypic hippocampal slice model is useful for studying delayed ischemic preconditioning that is maintained for hours or days after the preconditioning event.


Brain Research | 1998

Impaired expression of long-term potentiation in hippocampal slices 4 and 48 h following mild fluid-percussion brain injury in vivo

Thomas J. Sick; Miguel A. Perez-Pinzon; Zhen Zhou Feng

The effect of fluid percussion brain injury on hippocampal long-term potentiation (LTP) was investigated in hippocampal slices in vitro. Mild to moderate (1.7-2.1 atm) lateral fluid percussion head injury or sham operation was produced in rats 4 or 48 h prior to harvesting brain slices from the ipsilateral hippocampus. Field excitatory post-synaptic potentials (fEPSPs) were recorded in stratum radiatum of hippocampal subfield CA1 in response to electrical stimulation of the Schaffer collaterals. The initial slope of fEPSPs was used to investigate changes in synaptic strength prior to and following 100 or 200 Hz (1 s) tetanic stimulation. TBI significantly inhibited expression of LTP in hippocampal slices in vitro. Post-tetanus fEPSP slopes increased more than 100% in hippocampal slices from sham-operated animals but less than 50% in slices from rats following TBI. The data suggest that changes in functional synaptic plasticity in the hippocampus may contribute to cognitive disorders associated with TBI (traumatic brain injury). The data also indicate that TBI-induced effects on hippocampal LTP are robust and may be investigated in the hippocampal slice preparation in vitro.

Collaboration


Dive into the Thomas J. Sick's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph C. LaManna

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge