Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Stroke Res ; 11(2): 254-266, 2020 04.
Article in English | MEDLINE | ID: mdl-31250378

ABSTRACT

Hippocampal injury and cognitive impairments are common after cardiac arrest and stroke and do not have an effective intervention despite much effort. Therefore, we developed a new approach aimed at reversing synaptic dysfunction by targeting TRPM2 channels. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in mice was used to investigate cognitive deficits and the role of the calcium-permeable ion channel transient receptor potential-M2 (TRPM2) in ischemia-induced synaptic dysfunction. Our data indicates that absence (TRPM2-/-) or acute inhibition of TRPM2 channels with tatM2NX reduced hippocampal cell death in males only, but prevented synaptic plasticity deficits in both sexes. Remarkably, administration of tatM2NX weeks after injury reversed hippocampal plasticity and memory deficits. Finally, TRPM2-dependent activation of calcineurin-GSK3ß pathway contributes to synaptic plasticity impairments. These data suggest persistent TRPM2 activity following ischemia contributes to impairments of the surviving hippocampal network and that inhibition of TRPM2 channels at chronic time points may represent a novel strategy to improve functional recovery following cerebral ischemia that is independent of neuroprotection.


Subject(s)
Cognitive Dysfunction/physiopathology , Heart Arrest/complications , Hippocampus/physiopathology , Ischemia/complications , Neurons/physiology , TRPM Cation Channels/physiology , Animals , Calcineurin/physiology , Cardiopulmonary Resuscitation , Cognitive Dysfunction/etiology , Female , Glycogen Synthase Kinase 3 beta/physiology , Ischemia/physiopathology , Male , Mice, Knockout , Neuronal Plasticity , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
2.
J Cereb Blood Flow Metab ; 40(3): 588-599, 2020 03.
Article in English | MEDLINE | ID: mdl-30762478

ABSTRACT

Ischemic stroke is a leading cause of death worldwide and clinical data suggest that children may recover from stroke better than adults; however, supporting experimental data are lacking. We used our novel mouse model of experimental juvenile ischemic stroke (MCAO) to characterize age-specific cognitive dysfunction following ischemia. Juvenile and adult mice subjected to 45-min MCAO, and extracellular field recordings of CA1 neurons were performed to assess hippocampal synaptic plasticity changes after MCAO, and contextual fear conditioning was performed to evaluate memory and biochemistry used to analyze Nogo-A expression. Juvenile mice showed impaired synaptic plasticity seven days after MCAO, followed by full recovery by 30 days. Memory behavior was consistent with synaptic impairments and recovery after juvenile MCAO. Nogo-A expression increased in ipsilateral hippocampus seven days after MCAO compared to contralateral and sham hippocampus. Further, inhibition of Nogo-A receptors reversed MCAO-induced synaptic impairment in slices obtained seven days after juvenile MCAO. Adult MCAO-induced impairment of LTP was not associated with increased Nogo-A. This study demonstrates that stroke causes functional impairment in the hippocampus and recovery of behavioral and synaptic function is more robust in the young brain. Nogo-A receptor activity may account for the impairments seen following juvenile ischemic injury.


Subject(s)
Aging/metabolism , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Cognition , Nogo Proteins/metabolism , Signal Transduction , Stroke/metabolism , Aging/pathology , Animals , Brain Ischemia/pathology , CA1 Region, Hippocampal/pathology , Child , Disease Models, Animal , Humans , Memory , Mice , Neurons/metabolism , Neurons/pathology , Stroke/pathology
3.
J Cereb Blood Flow Metab ; 37(8): 3053-3064, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28168893

ABSTRACT

Cardiac arrest and cardiopulmonary resuscitation (CA/CPR) produce brain ischemia that results in cognitive and motor coordination impairments subsequent to injury of vulnerable populations of neurons, including cerebellar Purkinje neurons. To determine the effects of CA/CPR on plasticity in the cerebellum, we used whole cell recordings from Purkinje neurons to examine long-term depression (LTD) at parallel fiber (PF) synapses. Acute slices were prepared from adult male mice subjected to 8 min cardiac arrest at 1, 7, and 30 days after resuscitation. Concurrent stimulation of PF and climbing fibers (CFs) resulted in robust LTD of PF-evoked excitatory postsynaptic currents (EPSCs) in controls. LTD was absent in recordings obtained from mice subjected to CA/CPR, with no change in EPSC amplitude from baseline at any time point tested. AMPA and mGluR-mediated responses at the PF were not altered by CA/CPR. In contrast, CF-evoked NMDA currents were reduced following CA/CPR, which could account for the loss of LTD observed. A loss of GluN1 protein was observed following CA/CPR that was surprisingly not associated with changes in mRNA expression. These data demonstrate sustained impairments in synaptic plasticity in Purkinje neurons that survive the initial injury and which likely contribute to motor coordination impairments observed after CA/CPR.


Subject(s)
Cardiopulmonary Resuscitation , Excitatory Postsynaptic Potentials/physiology , Heart Arrest/physiopathology , Long-Term Synaptic Depression/physiology , Purkinje Cells , Animals , Disease Models, Animal , Heart Arrest/metabolism , Heart Arrest/pathology , Male , Mice, Inbred C57BL , Purkinje Cells/metabolism , Purkinje Cells/physiology , Receptors, Glutamate/metabolism
4.
Exp Neurol ; 283(Pt A): 151-6, 2016 09.
Article in English | MEDLINE | ID: mdl-27317297

ABSTRACT

INTRODUCTION: TRPM2 channels have been suggested to play a role in ischemic neuronal injury, specifically in males. A major hindrance to TRPM2 research has been the lack of specific TRPM2 inhibitors. The current study characterized the specificity and neuroprotective efficacy of a novel TRPM2 inhibitor. METHODS: Fluorescent calcium imaging (Fluo5F) was used to determine inhibitor efficacy of the TRPM2 peptide inhibitor (tat-M2NX) in HEK293 cells stably expressing hTRPM2. Adult (2-3months) and aged (18-20months) mice were subjected to 60min middle cerebral artery occlusion (MCAO) and injected with tat-M2NX, control scrambled peptide (tat-SCR) or clotrimazole (CTZ) either 20min prior or 3h after reperfusion. Infarct size was assessed using TTC staining. RESULTS: TRPM2 inhibition by tat-M2NX was observed by decreased Ca(2+) influx following H2O2 exposure human TRPM2 expressing cells. Male mice pre-treated with tat-M2NX had smaller infarct volume compared to tat-SCR. No effect of tat-M2NX on infarct size was observed in female mice. Importantly, male TRPM2(-/-) mice were not further protected by tat-M2NX, demonstrating selectivity of tat-M2NX. Administration of tat-M2NX 3h after reperfusion provided significant protection to males when analyzed at 24h or 4days after MCAO. Finally, we observed that tat-M2NX reduced ischemic injury in aged male mice. CONCLUSIONS: These data demonstrate the development of a new peptide inhibitor of TRPM2 channels that provides protection from ischemic stroke in young adult and aged male animals with a clinically relevant therapeutic window.


Subject(s)
Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , TRPM Cation Channels/chemistry , TRPM Cation Channels/metabolism , Age Factors , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Brain Ischemia/complications , Disease Models, Animal , Dose-Response Relationship, Drug , Female , HEK293 Cells/drug effects , HEK293 Cells/metabolism , Humans , Male , Mice , Mice, Knockout , Sex Factors , TRPM Cation Channels/genetics , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...