Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(14)2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37511013

ABSTRACT

In attention deficit hyperactivity disorder (ADHD), hyperactivity and impulsivity occur in response to delayed reward. Herein we report a novel animal model in which male Sprague-Dawley rats exposed to repeated hypoxic brain injury during the equivalent of extreme prematurity were ADHD-like hyperactive/impulsive in response to delayed reward and attentive at 3 months of age. Thus, a unique animal model of one of the presentations/subtypes of ADHD was discovered. An additional finding is that the repeated hypoxia rats were not hyperactive in the widely used open field test, which is not ADHD specific. Hence, it is recommended that ADHD-like hyperactivity and ADHD-like impulsivity, specifically in response to delayed reward, be a primary component in the design of future experiments that characterize potential animal models of ADHD, replacing open field testing of hyperactivity. Unknown is whether death and/or activity of midbrain dopaminergic neurons contributed to the ADHD-like hyperactivity/impulsivity detected after delayed reward. Hence, we stereologically measured the absolute number of dopaminergic neurons in four midbrain subregions and the average somal/nuclear volume of those neurons. Repeated hypoxia rats had a significant specific loss of dopaminergic neurons in the right ventral tegmental area (VTA) at 2 weeks of age and 18 months of age, providing new evidence of a site of pathology. No dopaminergic neuronal loss occurred in three other midbrain regions. Fewer VTA dopaminergic neurons correlated with increased ADHD-like hyperactivity and impulsivity. Novel early intervention therapies to rescue VTA dopaminergic neurons and potentially prevent ADHD-like hyperactivity/impulsivity can now be investigated.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Dopaminergic Neurons , Rats , Animals , Male , Dopaminergic Neurons/physiology , Rats, Sprague-Dawley , Ventral Tegmental Area , Reward , Impulsive Behavior , Hypoxia
2.
J Neurosci Res ; 101(2): 278-292, 2023 02.
Article in English | MEDLINE | ID: mdl-36412274

ABSTRACT

Stroke therapy has largely focused on preventing damage and encouraging repair outside the ischemic core, as the core is considered irreparable. Recently, several studies have suggested endogenous responses within the core are important for limiting the spread of damage and enhancing recovery, but the role of blood flow and capillary pericytes in this process is unknown. Using the Rose Bengal photothrombotic model of stroke, we illustrate blood vessels are present in the ischemic core and peri-lesional regions 2 weeks post stroke in male mice. A FITC-albumin gel cast of the vasculature revealed perfusion of these vessels, suggesting cerebral blood flow (CBF) may be partially present, without vascular leakage. The length of these vessels is significantly reduced compared to uninjured regions, but the average width is greater, suggesting they are either larger vessels that survived the initial injury, smaller vessels that have expanded in size (i.e., arteriogenesis), or that neovascularization begins with larger vessels. Concurrently, we observed an increase in platelet-derived growth factor receptor beta (PDGFRß, a marker of pericytes) expression within the ischemic core in two distinct patterns, one which resembles pericyte-derived fibrotic scarring at the edge of the core, and one which is vessel associated and may represent blood vessel recovery. We find little evidence for dividing cells on these intralesional blood vessels 2 weeks post stroke. Our study provides evidence flow is present in PDGFRß-positive vessels in the ischemic core 2 weeks post stroke. We hypothesize intralesional CBF is important for limiting injury and for encouraging endogenous repair following cerebral ischemia.


Subject(s)
Rose Bengal , Serum Albumin , Male , Mice , Animals
3.
Biomed Pharmacother ; 156: 113895, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36274464

ABSTRACT

Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα) is a potential target for acute neuroprotection due to its key role in physiological and pathological glutamate signaling. The hub domain organizes the CaMKII holoenzyme into large oligomers, and additional functional effects on holoenzyme activation have lately emerged. We recently reported that compounds related to the proposed neuromodulator γ-hydroxybutyrate (GHB) selectively bind to the CaMKIIα hub domain and increase hub thermal stabilization, which is believed to have functional consequences and to mediate neuroprotection. However, the detailed molecular mechanism is unknown. In this study, we functionally characterize the novel and brain permeable GHB analog (E)-2-(5-hydroxy-2-phenyl-5,7,8,9-tetrahydro-6H-benzo[7]annulen-6-ylidene)acetic acid (Ph-HTBA). Administration of a single dose of Ph-HTBA at a clinically relevant time point (3-6 h after photothrombotic stroke) promotes neuroprotection with a superior effect at low doses compared to the smaller GHB analog 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA). In contrast to HOCPCA, Ph-HTBA reduces Ca2+-stimulated CaMKIIα Thr286 autophosphorylation in primary cortical neurons and substrate phosphorylation of recombinant CaMKIIα, potentially contributing to its neuroprotective effect. Supported by previous in silico docking studies, we suggest that Ph-HTBA makes distinct molecular interactions with the hub cavity, which may contribute to its differential functional profile and superior neuroprotective effect compared to HOCPCA. Together, this highlights Ph-HTBA as a promising tool to study hub functionality, but also as a good candidate for clinical development.


Subject(s)
Ischemic Stroke , Neuroprotective Agents , Sodium Oxybate , Humans , Ligands , Sodium Oxybate/metabolism , Neuroprotection , Neuroprotective Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Holoenzymes
4.
Lipids ; 57(1): 17-31, 2022 01.
Article in English | MEDLINE | ID: mdl-34751447

ABSTRACT

N-acylethanolamines (NAE, also called ethanolamides) are significant lipid signaling molecules with anti-inflammatory, pain-relieving, cell-protective, and anticancer properties. Here, we present the use of a hitherto unreported group of Δ3-NAE and also some Δ4- and Δ5-NAE, in in vitro and in vivo assays to gain a better understanding of their structure-bioactivity relationships. We have developed an efficient synthetic method to rapidly produce novel unlabeled and 13 C-labeled Δ3-NAE (NAE-18:5n-3, NAE-18:4n-6) and Δ4-NAE (NAE-22:5n-6). The new NAE with shorter carbon backbone structures confers greater neuroprotection than their longer carbon backbone counterparts, including anandamide (Δ5-NAE-20:4n-6) in a focal ischemia mouse model of stroke. This study highlights structure-dependent protective effects of new NAE following focal ischemia, in which some of the new NAE, administered intranasally, lead to significantly reduced infarct volume and improved recovery of limb use. The relative affinity of the new NAE toward cannabinoid receptors was assessed against anandamide, NAE-22:6n-3 and NAE-20:5n-3, which are known cannabinoid receptor ligands with high-binding constants. Among the newly synthesized NAE, Δ4-NAE-22:5n-6 shows the greatest relative affinity to cannabinoid receptors hCB1 and hCB2 , and inhibition of cyclic adenosine monophosphate activity through hCB2 compared to anandamide.


Subject(s)
Neuroprotection , Stroke , Animals , Ethanolamines , Mice , Receptors, Cannabinoid
5.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34210784

ABSTRACT

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Subject(s)
Diazepam Binding Inhibitor/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Neurons/drug effects , Neuropeptides/therapeutic use , Peptide Fragments/therapeutic use , Receptors, GABA-A/drug effects , Stroke/drug therapy , Adult , Animals , Astrocytes/metabolism , Cortical Spreading Depression/physiology , Diazepam Binding Inhibitor/deficiency , Diazepam Binding Inhibitor/physiology , Drug Implants , Evoked Potentials, Somatosensory , Female , GABA-A Receptor Agonists/pharmacology , Humans , Hydrogels , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/etiology , Light , Mice , Mice, Inbred C57BL , N-Methylaspartate/toxicity , Neurons/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Patch-Clamp Techniques , Peptide Fragments/deficiency , Peptide Fragments/physiology , Rats , Rose Bengal/radiation effects , Rose Bengal/toxicity , Single-Blind Method , Stroke/etiology
6.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34330837

ABSTRACT

Ca2+/calmodulin-dependent protein kinase II alpha subunit (CaMKIIα) is a key neuronal signaling protein and an emerging drug target. The central hub domain regulates the activity of CaMKIIα by organizing the holoenzyme complex into functional oligomers, yet pharmacological modulation of the hub domain has never been demonstrated. Here, using a combination of photoaffinity labeling and chemical proteomics, we show that compounds related to the natural substance γ-hydroxybutyrate (GHB) bind selectively to CaMKIIα. By means of a 2.2-Å x-ray crystal structure of ligand-bound CaMKIIα hub, we reveal the molecular details of the binding site deep within the hub. Furthermore, we show that binding of GHB and related analogs to this site promotes concentration-dependent increases in hub thermal stability believed to alter holoenzyme functionality. Selectively under states of pathological CaMKIIα activation, hub ligands provide a significant and sustained neuroprotection, which is both time and dose dependent. This is demonstrated in neurons exposed to excitotoxicity and in a mouse model of cerebral ischemia with the selective GHB analog, HOCPCA (3-hydroxycyclopent-1-enecarboxylic acid). Together, our results indicate a hitherto unknown mechanism for neuroprotection by a highly specific and unforeseen interaction between the CaMKIIα hub domain and small molecule brain-penetrant GHB analogs. This establishes GHB analogs as powerful tools for investigating CaMKII neuropharmacology in general and as potential therapeutic compounds for cerebral ischemia in particular.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Sodium Oxybate/metabolism , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carboxylic Acids/pharmacology , Crystallography, X-Ray , Cyclopentanes/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , HEK293 Cells , Humans , Neuroprotection , Protein Binding , Protein Domains , Signal Transduction
7.
Transl Stroke Res ; 12(1): 72-86, 2021 02.
Article in English | MEDLINE | ID: mdl-32253702

ABSTRACT

The extracellular matrix fragment perlecan domain V is neuroprotective and functionally restorative following experimental stroke. As neurogenesis is an important component of chronic post-stroke repair, and previous studies have implicated perlecan in developmental neurogenesis, we hypothesized that domain V could have a broad therapeutic window by enhancing neurogenesis after stroke. We demonstrated that domain V is chronically increased in the brains of human stroke patients, suggesting that it is present during post-stroke neurogenic periods. Furthermore, perlecan deficient mice had significantly less neuroblast precursor cells after experimental stroke. Seven-day delayed domain V administration enhanced neurogenesis and restored peri-infarct excitatory synaptic drive to neocortical layer 2/3 pyramidal neurons after experimental stroke. Domain V's effects were inhibited by blockade of α2ß1 integrin, suggesting the importance of α2ß1 integrin to neurogenesis and domain V neurogenic effects. Our results demonstrate that perlecan plays a previously unrecognized role in post-stroke neurogenesis and that delayed DV administration after experimental stroke enhances neurogenesis and improves recovery in an α2ß1 integrin-mediated fashion. We conclude that domain V is a clinically relevant neuroprotective and neuroreparative novel stroke therapy with a broad therapeutic window.


Subject(s)
Brain/metabolism , Heparan Sulfate Proteoglycans/biosynthesis , Neurogenesis/physiology , Neuroprotection/physiology , Stroke/metabolism , Animals , Brain/drug effects , Brain/pathology , Cells, Cultured , Heparan Sulfate Proteoglycans/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Neuroprotection/drug effects , Organ Culture Techniques , Protein Domains , Stroke/pathology , Stroke/prevention & control
8.
Front Neurosci ; 13: 1133, 2019.
Article in English | MEDLINE | ID: mdl-31736685

ABSTRACT

Inflammatory processes are known to contribute to tissue damage in the central nervous system (CNS) across a broad range of neurological conditions, including stroke. Gamma amino butyric acid (GABA), the main inhibitory neurotransmitter in the CNS, has been implicated in modulating peripheral immune responses by acting on GABA A receptors on antigen-presenting cells and lymphocytes. Here, we investigated the effects and mechanism of action of the delta-selective compound, DS2, to improve stroke recovery and modulate inflammation. We report a decrease in nuclear factor (NF)-κB activation in innate immune cells over a concentration range in vitro. Following a photochemically induced motor cortex stroke, treatment with DS2 at 0.1 mg/kg from 1 h post-stroke significantly decreased circulating tumor necrosis factor (TNF)-α, interleukin (IL)-17, and IL-6 levels, reduced infarct size and improved motor function in mice. Free brain concentrations of DS2 were found to be lower than needed for robust modulation of central GABA A receptors and were not affected by the presence and absence of elacridar, an inhibitor of both P-glycoprotein and breast cancer resistance protein (BCRP). Finally, as DS2 appears to dampen peripheral immune activation and only shows limited brain exposure, we assessed the role of DS2 to promote functional recovery after stroke when administered from 3-days after the stroke. Treatment with DS2 from 3-days post-stroke improved motor function on the grid-walking, but not on the cylinder task. These data highlight the need to further develop subunit-selective compounds to better understand change in GABA receptor signaling pathways both centrally and peripherally. Importantly, we show that GABA compounds such as DS2 that only shows limited brain exposure can still afford significant protection and promote functional recovery most likely via modulation of peripheral immune cells and could be given as an adjunct treatment.

9.
Neuromolecular Med ; 21(4): 401-413, 2019 12.
Article in English | MEDLINE | ID: mdl-31313065

ABSTRACT

Frontal infarcts can produce cognitive impairments that affect an individual's ability to function in everyday life. However, the precise types of deficits, and their underlying mechanisms, are not well-understood. Here we used a prefrontal photothrombotic stroke model in C57BL/6J mice to characterise specific cognitive changes that occur in the 6 weeks post-stroke. Behavioural experiments were paired with in vivo electrophysiology to assess whether changes in oscillatory communication between the prefrontal cortex (PFC) and the hippocampus (HPC) mirrored any observed behavioural changes. We found that mice in the stroke group exhibited a delayed onset impairment in tasks of spatial working memory (object location recognition and Y-maze) and that this correlated with reduced PFC-HPC theta band coherence (5-12 Hz) during the task. In the open field, mice in the stroke group exhibited hyperactivity as compared to controls, and stroke animals also exhibited significantly higher beta band activity (13-30 Hz) in the PFC and the HPC. Taken together our results suggest that infarcts in the PFC result in PFC-HPC oscillatory communication changes in the theta and beta bands, correlating with altered performance in spatial memory and open field tasks respectively. Of particular interest, early open field changes in PFC beta band power post-stroke correlated to later-stage spatial memory impairments, highlighting this as a potential biomarker for detecting when spatial memory impairments are likely to occur.


Subject(s)
Hippocampus/physiopathology , Maze Learning/physiology , Memory Disorders/etiology , Prefrontal Cortex/physiopathology , Stroke/complications , Animals , Beta Rhythm , Electrodes, Implanted , Exploratory Behavior/physiology , Male , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Neural Pathways/physiopathology , Open Field Test/physiology , Random Allocation , Recognition, Psychology/physiology , Stroke/pathology , Stroke/physiopathology , Stroke/psychology , Theta Rhythm
10.
Neural Plast ; 2019: 1460890, 2019.
Article in English | MEDLINE | ID: mdl-31191635

ABSTRACT

Stroke remains a leading cause of disability worldwide. Recently, we have established an animal model of stroke that results in delayed impairment in spatial memory, allowing us to better investigate cognitive deficits. Young and aged brains show different recovery profiles after stroke; therefore, we assessed aged-related differences in poststroke cognition. As neurotrophic support diminishes with age, we also investigated the involvement of brain-derived neurotrophic factor (BDNF) in these differences. Young (3-6 months old) and aged (16-21 months old) mice were trained in operant touchscreen chambers to complete a visual pairwise discrimination (VD) task. Stroke or sham surgery was induced using the photothrombotic model to induce a bilateral prefrontal cortex stroke. Five days poststroke, an additional cohort of aged stroke animals were treated with intracerebral hydrogels loaded with the BDNF decoy, TrkB-Fc. Following treatment, animals underwent the reversal and rereversal task to identify stroke-induced cognitive deficits at days 17 and 37 poststroke, respectively. Assessment of sham animals using Cox regression and log-rank analyses showed aged mice exhibit an increased impairment on VD reversal and rereversal learning compared to young controls. Stroke to young mice revealed no impairment on either task. In contrast, stroke to aged mice facilitated a significant improvement in reversal learning, which was dampened in the presence of the BDNF decoy, TrkB-Fc. In addition, aged stroke control animals required significantly less consecutive days and correction trials to master the reversal task, relative to aged shams, an effect dampened by TrkB-Fc. Our findings support age-related differences in recovery of cognitive function after stroke. Interestingly, aged stroke animals outperformed their sham counterparts, suggesting reopening of a critical window for recovery that is being mediated by BDNF.


Subject(s)
Cognition/physiology , Recovery of Function/physiology , Reversal Learning/physiology , Stroke/psychology , Animals , Discrimination Learning/physiology , Disease Models, Animal , Male , Mice , Receptor, trkB/metabolism , Stroke/metabolism
11.
J Cereb Blood Flow Metab ; 39(1): 74-88, 2019 01.
Article in English | MEDLINE | ID: mdl-29160736

ABSTRACT

Ischemic stroke triggers an elevation in tonic GABA inhibition that impairs the ability of the brain to form new structural and functional cortical circuits required for recovery. This stroke-induced increase in tonic inhibition is caused by impaired GABA uptake via the glial GABA transporter GAT3, highlighting GAT3 as a novel target in stroke recovery. Using a photothrombotic stroke mouse model, we show that GAT3 protein levels are decreased in peri-infarct tissue from 6 h to 42 days post-stroke. Prior studies have shown that GAT substrates can increase GAT surface expression. Therefore, we aimed to assess whether the GAT3 substrate, L-isoserine, could increase post-stroke functional recovery. L-Isoserine (38 µM or 380 µM) administered directly into the infarct from day 5 to 32 post-stroke, significantly increased motor performance in the grid-walking and cylinder tasks in a concentration-dependent manner, without affecting infarct volumes. Additionally, L-isoserine induced a lasting increase in GAT3 expression in peri-infarct regions accompanied by a small decrease in GFAP expression. This study is the first to show that a GAT3 substrate can increase GAT3 expression and functional recovery after focal ischemic stroke following a delayed long-term treatment. We propose that enhancing GAT3-mediated uptake dampens tonic inhibition and promotes functional recovery after stroke.


Subject(s)
Brain Ischemia/drug therapy , GABA Plasma Membrane Transport Proteins/biosynthesis , Recovery of Function/drug effects , Serine/analogs & derivatives , Stroke/drug therapy , Animals , Brain Ischemia/physiopathology , Dose-Response Relationship, Drug , GABA Plasma Membrane Transport Proteins/genetics , Glial Fibrillary Acidic Protein/biosynthesis , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Serine/pharmacology , Serine/therapeutic use , Stroke/physiopathology , Up-Regulation/drug effects
12.
J Cereb Blood Flow Metab ; 39(7): 1266-1282, 2019 07.
Article in English | MEDLINE | ID: mdl-29376464

ABSTRACT

Tonic inhibitory currents, mediated by extrasynaptic GABAA receptors, are elevated at a delay following stroke. Flavonoids minimise the extent of cellular damage following stroke, but little is known about their mode of action. We demonstrate that the flavonoid, 2'-methoxy-6-methylflavone (0.1-10 µM; 2'MeO6MF), increases GABAA receptor tonic currents presumably via δ-containing GABAA receptors. Treatment with 2'MeO6MF 1-6 h post focal ischaemia dose dependently decreases infarct volume and improves functional recovery. The effect of 2'MeO6MF was attenuated in δ-/- mice, indicating that the effects of the flavonoid were mediated via δ-containing GABAA receptors. Further, as flavonoids have been shown to have multiple modes of action, we investigated the anti-inflammatory effects of 2'MeO6MF. Using a macrophage cell line, we show that 2'MeO6MF can dampen an LPS-induced elevation in NFkB activity. Assessment of vehicle-treated stroke animals revealed a significant increase in circulating IL1ß, TNFα and IFγ levels. Treatment with 2'MeO6MF dampened the stroke-induced increase in circulating cytokines, which was blocked in the presence of the pan-AKT inhibitor, GSK690693. These studies support the hypothesis that compounds that potentiate tonic inhibition via δ-containing GABAA receptors soon after stroke can afford neuroprotection.


Subject(s)
Brain Ischemia/drug therapy , Flavones/administration & dosage , GABA Modulators/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Brain/metabolism , Disease Models, Animal , Flavones/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Stroke/drug therapy , Synaptic Potentials/drug effects , Synaptic Potentials/physiology
13.
Neuroscience ; 393: 185-195, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30059704

ABSTRACT

Despite the fact that approximately 80% of strokes occur in those aged over 60 years, many pre-clinical stroke studies have been conducted in younger adult rodents, raising debate about translation and generalizability of these results. We were interested in potential age differences in stroke-induced secondary neurodegeneration (SND). SND involves the death of neurons in areas remote from, but connected to, the site of infarction, as well as glial disturbances. Here we investigated potential differences in key parameters of SND in the thalamus, a major site of post-stroke SND. Protein expression profiles in young adult (2-4 months) and aged (22-23 months) mice were analyzed 28 days after a cortical stroke. Our results show that age reduced the expression of synaptic markers (PSD 95, Synapsin1) and increased Amyloid ß oligomer accumulation after stroke. Protein expression of several markers of glial activity remained relatively stable across age groups post-stroke. We have identified that age exacerbates the severity of SND after stroke. Our results, however, do not support a view that microglia or astrocytes are the main contributors to the enhanced severity of SND in aged mice.


Subject(s)
Age Factors , Microglia/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Stroke/pathology , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Male , Mice, Inbred C57BL , Neuroglia/metabolism , Thalamus/metabolism
14.
J Appl Physiol (1985) ; 125(3): 862-869, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29856262

ABSTRACT

It is known that high blood pressure variability (BPV) in acute ischemic stroke is associated with adverse outcomes, yet there are no therapeutic treatments to reduce BPV. Studies have found increasing nitric oxide (NO) bioavailability improves neurological function following stroke, but whether dietary nitrate supplementation could reduce BPV remains unknown. We investigated the effects of dietary nitrate supplementation on heart rate (HR), blood pressure (BP), and beat-to-beat BPV using wireless telemetry in a rat model of distal middle cerebral artery occlusion. Blood pressure variability was characterized by spectral power analysis in the low frequency (LF; 0.2-0.6 Hz) range prestroke and during the 7 days poststroke in a control group ( n = 8) and a treatment group ( n = 8, 183 mg/l sodium nitrate in drinking water). Dietary nitrate supplementation moderately reduced systolic BPV in the LF range by ~11% compared with the control group ( P = 0.03), while resting BP and HR were not different between the two groups ( P = 0.28 and 0.33, respectively). Despite systolic BPV being reduced with dietary nitrate, we found no difference in infarct volumes between the treatment and the control groups (1.59 vs. 1.62 mm3, P = 0.86). These findings indicate that dietary nitrate supplementation is effective in reducing systolic BPV following stroke without affecting absolute BP. In light of mounting evidence linking increased BPV with poor stroke patient outcome, our data support the role of dietary nitrate as an adjunct treatment following ischemic stroke. NEW & NOTEWORTHY Using a rat model of stroke, we found that dietary nitrate supplementation reduced low frequency blood pressure fluctuations following stroke without affecting absolute blood pressure values. Since blood pressure fluctuations are associated with poor clinical outcome in stroke patients, our findings indicate that dietary nitrate could be an effective strategy for reducing blood pressure fluctuations, which could help reduce stroke severity and improve patient recovery.


Subject(s)
Blood Pressure/drug effects , Dietary Supplements , Infarction, Middle Cerebral Artery/drug therapy , Nitrates/therapeutic use , Animals , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Heart Rate/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Stroke/drug therapy , Stroke/physiopathology
15.
J Neurophysiol ; 120(3): 1143-1152, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29873614

ABSTRACT

Serial-order behavior is the ability to complete a sequence of responses in a predetermined order to achieve a reward. In birds, serial-order behavior is thought to be impaired by damage to the nidopallium caudolaterale (NCL). In the current study, we examined the role of the NCL in serial-order behavior by training pigeons on a 4-item serial-order task and a go/no-go discrimination task. Following training, pigeons received infusions of 1 µl of either tetrodotoxin (TTX) or saline. Saline infusions had no impact on serial-order behavior, whereas TTX infusions resulted in a significant decrease in performance. The serial-order impairments, however, were not the result of any specific error at any specific list item. With respect to the go/no-go discrimination task, saline infusions also had no impact on performance, whereas TTX infusions impaired pigeons' discrimination abilities. Given the impairments on the go/no-go discrimination task, which does not require processing of serial-order information, we tentatively conclude that damage to the NCL does not impair serial-order behavior per se, but rather results in a more generalized impairment that may impact performance across a range of tasks. NEW & NOTEWORTHY We examined the role of the nidopallium caudolaterale (NCL) in serial-order behavior by training pigeons on a 4-item serial-order task and selectively inhibiting the region with TTX. Although TTX infusions did impair serial-order behavior, the pattern of the deficit, plus the fact that TTX also impaired performance on a task without a serial-order component, indicates that inactivation of NCL causes impairments in reward processing or inhibition rather than serial-order behavior.


Subject(s)
Discrimination Learning/physiology , Globus Pallidus/physiology , Serial Learning/physiology , Animals , Columbidae , Conditioning, Operant , Reward
16.
Stroke ; 49(3): 700-709, 2018 03.
Article in English | MEDLINE | ID: mdl-29382802

ABSTRACT

BACKGROUND AND PURPOSE: Human amnion epithelial cells (hAECs) are nonimmunogenic, nontumorigenic, anti-inflammatory cells normally discarded with placental tissue. We reasoned that their profile of biological features, wide availability, and the lack of ethical barriers to their use could make these cells useful as a therapy in ischemic stroke. METHODS: We tested the efficacy of acute (1.5 hours) or delayed (1-3 days) poststroke intravenous injection of hAECs in 4 established animal models of cerebral ischemia. Animals included young (7-14 weeks) and aged mice (20-22 months) of both sexes, as well as adult marmosets of either sex. RESULTS: We found that hAECs administered 1.5 hours after stroke in mice migrated to the ischemic brain via a CXC chemokine receptor type 4-dependent mechanism and reduced brain inflammation, infarct development, and functional deficits. Furthermore, if hAECs administration was delayed until 1 or 3 days poststroke, long-term functional recovery was still augmented in young and aged mice of both sexes. We also showed proof-of-principle evidence in marmosets that acute intravenous injection of hAECs prevented infarct development from day 1 to day 10 after stroke. CONCLUSIONS: Systemic poststroke administration of hAECs elicits marked neuroprotection and facilitates mechanisms of repair and recovery.


Subject(s)
Amnion/transplantation , Epithelial Cells/transplantation , Neuroprotection , Stroke/therapy , Animals , Callithrix , Disease Models, Animal , Female , Heterografts , Humans , Male , Mice , Stroke/metabolism , Stroke/pathology
17.
J Cereb Blood Flow Metab ; 38(1): 166-173, 2018 01.
Article in English | MEDLINE | ID: mdl-29148909

ABSTRACT

Brain ischemia triggers excitotoxicity and cell death, yet no neuroprotective drugs have made it to the clinic. While enhancing GABAergic signaling to counterbalance excitotoxicity has shown promise in animal models, clinical studies have failed. Blockade of GABA transporters (GATs) offers an indirect approach to increase GABA inhibition to lower the excitation threshold of neurons. Among the GATs, GAT1 is known to promote neuroprotection, while the protective role of the extrasynaptic transporters GAT3 and BGT1 is elusive. A focal lesion was induced in the motor cortex in two to four-month-old C57BL/6 J male mice by photothrombosis. The GAT1 inhibitor, tiagabine (1 and 10 mg/kg), the GAT2/3 inhibitor, ( S)-SNAP-5114 (5 and 30 mg/kg) and the GAT1/BGT1 inhibitor, EF-1502 (1 and 10 mg/kg) were given i.p. 1 and 6 h post-stroke to assess their impact on infarct volume and motor performance seven days post-stroke. One mg/kg tiagabine improved motor performance, while 10 mg/kg tiagabine, ( S)-SNAP-5114 and EF-1502 had no effect. None of the compounds affected infarct volume. Interestingly, treatment with tiagabine induced seizures and ( S)-SNAP-5114 led to increased mortality. Although we show that tiagabine can promote protection, our findings indicate that caution should be had when using GAT1 and GAT3 inhibitors for conditions of brain ischemia.


Subject(s)
Brain Ischemia/pathology , GABA Antagonists/pharmacology , Recovery of Function/drug effects , Animals , Brain Ischemia/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology
18.
Sci Rep ; 6: 21896, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902390

ABSTRACT

Although recovery following a stroke is limited, undamaged neurons under the right conditions can establish new connections and take on-board lost functions. Sonic hedgehog (Shh) signaling is integral for developmental axon growth, but its role after injury has not been fully examined. To investigate the effects of Shh on neuronal sprouting after injury, we used an in vitro model of glial scar, whereby cortical astrocytes were mechanically traumatized to mimic reactive astrogliosis observed after stroke. This mechanical trauma impaired neurite outgrowth from post-natal cortical neurons plated on top of reactive astrocytes. Addition of Shh to the media, however, resulted in a concentration-dependent increase in neurite outgrowth. This response was inhibited by cyclopamine and activated by oxysterol 20(S)-hydroxycholesterol, both of which modulate the activity of the Shh co-receptor Smoothened (Smo), demonstrating that Shh-mediated neurite outgrowth is Smo-dependent. In addition, neurite outgrowth was not associated with an increase in Gli-1 transcription, but could be inhibited by PP2, a selective inhibitor of Src family kinases. These results demonstrate that neurons exposed to the neurite growth inhibitory environment associated with a glial scar can be stimulated by Shh, with signaling occurring through a non-canonical pathway, to overcome this suppression and stimulate neurite outgrowth.


Subject(s)
Astrocytes/drug effects , Gliosis/metabolism , Hedgehog Proteins/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Coculture Techniques , Gene Expression Regulation , Gliosis/genetics , Gliosis/pathology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hydroxycholesterols/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/cytology , Neurons/metabolism , Pressure , Primary Cell Culture , Pyrimidines/pharmacology , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stress, Mechanical , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
19.
Mol Cell Neurosci ; 68: 56-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25828540

ABSTRACT

Perinatal hypoxia-ischemia is a major cause of striatal injury and may lead to cerebral palsy. This study investigated whether delayed administration of bone marrow-derived mesenchymal stem cells (MSCs), at one week after neonatal rat hypoxia-ischemia, was neurorestorative of striatal medium-spiny projection neurons and improved motor function. The effect of a subcutaneous injection of a high-dose, or a low-dose, of MSCs was investigated in stereological studies. Postnatal day (PN) 7 pups were subjected to hypoxia-ischemia. At PN14, pups received treatment with either MSCs or diluent. A subset of high-dose pups, and their diluent control pups, were also injected intraperitoneally with bromodeoxyuridine (BrdU), every 24h, on PN15, PN16 and PN17. This permitted tracking of the migration and survival of neuroblasts originating from the subventricular zone into the adjacent injured striatum. Pups were euthanized on PN21 and the absolute number of striatal medium-spiny projection neurons was measured after immunostaining for DARPP-32 (dopamine- and cAMP-regulated phosphoprotein-32), double immunostaining for BrdU and DARPP-32, and after cresyl violet staining alone. The absolute number of striatal immunostained calretinin interneurons was also measured. There was a statistically significant increase in the absolute number of DARPP-32-positive, BrdU/DARPP-32-positive, and cresyl violet-stained striatal medium-spiny projection neurons, and fewer striatal calretinin interneurons, in the high-dose mesenchymal stem cell (MSC) group compared to their diluent counterparts. A high-dose of MSCs restored the absolute number of these neurons to normal uninjured levels, when compared with previous stereological data on the absolute number of cresyl violet-stained striatal medium-spiny projection neurons in the normal uninjured brain. For the low-dose experiment, in which cresyl violet-stained striatal medium-spiny neurons alone were measured, there was a lower statistically significant increase in their absolute number in the MSC group compared to their diluent controls. Investigation of behavior in another cohort of animals showed that delayed administration of a high-dose of bone marrow-derived MSCs, at one week after neonatal rat hypoxia-ischemia, improved motor function on the cylinder test. Thus, delayed therapy with a high- or low-dose of adult MSCs, at one week after injury, is effective in restoring the loss of striatal medium-spiny projection neurons after neonatal rat hypoxia-ischemia and a high-dose of MSCs improved motor function.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Corpus Striatum/pathology , Hypoxia-Ischemia, Brain , Mesenchymal Stem Cells/physiology , Motor Skills Disorders/therapy , Neurons/physiology , Age Factors , Animals , Animals, Newborn , Antigens, CD/metabolism , Body Weight , Calbindin 2/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/therapy , Lateral Ventricles/cytology , Male , Motor Skills Disorders/etiology , Neurogenesis , Rats , Rats, Sprague-Dawley , Time Factors
20.
J Cereb Blood Flow Metab ; 35(8): 1272-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25757752

ABSTRACT

Cerebral ischemia results in damage to neuronal circuits and lasting impairment in function. We have previously reported that stimulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors with the ampakine, CX1837, increases brain-derived neurotrophic factor (BDNF) levels and affords significant motor recovery after stroke in young mice. Here, we investigated whether administration of CX1837 in aged (24 months old) mice was equally effective. In a model of focal ischemia, administration of CX1837 from 5 days after stroke resulted in a small gain of motor function by week 6 after stroke. Mice that received a local delivery of BDNF via hydrogel implanted into the stroke cavity also showed a small gain of function from 4 to 6 weeks after stroke. Combining both treatments, however, resulted in a marked improvement in motor function from 2 weeks after insult. Assessment of peri-infarct tissue 2 weeks after stroke revealed a significant increase in p-AKT and p-CREB after the combined drug treatment. Using the pan-AKT inhibitor, GSK-690693, or deletion of CREB from forebrain neurons using the CREB-flox/CAMKii-cre mice, we were able to block the recovery of motor function. These data suggest that combined CX1837 and local delivery of BDNF are required to achieve maximal functional recovery after stroke in aged mice, and is occurring via the AKT-GSK3-CREB signaling pathway.


Subject(s)
Brain Ischemia , Brain-Derived Neurotrophic Factor/pharmacology , Cyclic AMP Response Element-Binding Protein , Propionates/pharmacology , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Stroke , Animals , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recovery of Function/drug effects , Signal Transduction/genetics , Stroke/drug therapy , Stroke/genetics , Stroke/metabolism , Stroke/pathology , Stroke/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...