Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
J Psychiatr Res ; 164: 192-201, 2023 08.
Article in English | MEDLINE | ID: mdl-37356352

ABSTRACT

Lithium carbonate (LiCO) is a mainstay therapeutic for the prevention of mood-episode recurrences in bipolar disorder (BD). Unfortunately, its narrow therapeutic index is associated with complications that may lead to treatment non-compliance. Intriguingly, lithium orotate (LiOr) is suggested to possess unique uptake characteristics that would allow for reduced dosing and mitigation of toxicity concerns. We hypothesized that due to differences in pharmacokinetics, LiOr is more potent with reduced adverse effects. Dose responses were established for LiOr and LiCO in male and female mice using an amphetamine-induced hyperlocomotion (AIH) model; AIH captures manic elements of BD and is sensitive to a dose-dependent lithium blockade. LiCO induced a partial block of AIH at doses of 15 mg/kg in males and 20 mg/kg in females. In contrast, LiOr elicited a near complete blockade at concentrations of just 1.5 mg/kg in both sexes, indicating improved efficacy and potency. Prior application of organic anion transport inhibitors, or inhibition of orotate uptake into the pentose pathway, completely blocked the effects of LiOr on AIH while sparing LiCO effects, confirming differences in transport and compartmentalization between the two compounds. Next, the relative toxicities of LiOr and LiCO were contrasted after 14 consecutive daily administrations. LiCO, but not LiOr, elicited polydipsia in both sexes, elevated serum creatinine levels in males, and increased serum TSH expression in females. LiOr demonstrates superior efficacy, potency, and tolerability to LiCO in both male and female mice because of select transport-mediated uptake and pentose pathway incorporation.


Subject(s)
Bipolar Disorder , Lithium Carbonate , Male , Female , Mice , Animals , Lithium Carbonate/adverse effects , Mania/chemically induced , Mania/drug therapy , Bipolar Disorder/drug therapy , Lithium/therapeutic use , Amphetamine/therapeutic use , Disease Models, Animal , Antimanic Agents/pharmacology
2.
Brain Behav ; 11(8): e2262, 2021 08.
Article in English | MEDLINE | ID: mdl-34196467

ABSTRACT

Bipolar disorder (BD) poses a significant public health concern, with roughly one-quarter of sufferers attempting suicide. BD is characterized by manic and depressive mood cycles, the recurrence of which can be effectively curtailed through lithium therapy. Unfortunately, the most frequently employed lithium salt, lithium carbonate (Li2 CO3 ), is associated with a host of adverse health outcomes following chronic use: these unwanted effects range from relatively minor inconveniences (e.g., polydipsia and polyuria) to potentially major complications (e.g., hypothyroidism and/or renal impairment). As these undesirable effects can limit patient compliance, an alternative lithium compound with a lesser toxicity profile would dramatically improve treatment efficacy and outcomes. Lithium orotate (LiC5 H3 N2 O4 ; henceforth referred to as LiOr), a compound largely abandoned since the late 1970s, may represent such an alternative. LiOr is proposed to cross the blood-brain barrier and enter cells more readily than Li2 CO3 , which will theoretically allow for reduced dosage requirements and ameliorated toxicity concerns. This review addresses the controversial history of LiOr, complete with discussions of experimental and clinical efficacy, putative mechanisms of action, adverse effects, and its potential future in therapy.


Subject(s)
Bipolar Disorder , Lithium , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Humans , Lithium/therapeutic use , Lithium Compounds/adverse effects , Organometallic Compounds
3.
Eur J Neurosci ; 54(8): 6948-6959, 2021 10.
Article in English | MEDLINE | ID: mdl-33190318

ABSTRACT

Catecholamine neurons of the locus coeruleus (LC) in the dorsal pontine tegmentum innervate the entire neuroaxis, with signaling actions implicated in the regulation of attention, arousal, sleep-wake cycle, learning, memory, anxiety, pain, mood, and brain metabolism. The co-release of norepinephrine (NE) and dopamine (DA) from LC terminals in the hippocampus plays a role in all stages of hippocampal-memory processing. This catecholaminergic regulation modulates the encoding, consolidation, retrieval, and reversal of hippocampus-based memory. LC neurons in awake animals have two distinct firing modes: tonic firing (explorative) and phasic firing (exploitative). These two firing modes exert different modulatory effects on post-synaptic dendritic spines. In the hippocampus, the firing modes regulate long-term potentiation (LTP) and long-term depression, which differentially regulate the mRNA expression and transcription of plasticity-related proteins (PRPs). These proteins aid in structural alterations of dendritic spines, that is, structural long-term potentiation (sLTP), via expansion and structural long-term depression (sLTD) via contraction of post-synaptic dendritic spines. Given the LC's role in all phases of memory processing, the degeneration of 50% of the LC neuron population occurring in Alzheimer's disease (AD) is a clinically relevant aspect of disease pathology. The loss of catecholaminergic regulation contributes to dysfunction in memory processes along with impaired functions associated with attention and task completion. The multifaceted role of the LC in memory and general task performance and the close correlation of LC degeneration with neurodegenerative disease progression together implicate it as a target for new clinical assessment tools.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Animals , Locus Coeruleus , Long-Term Potentiation , Memory
4.
Physiol Behav ; 229: 113287, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33316294

ABSTRACT

Diabetes is a prevalent metabolic disorder that has long been associated with changes in different regions of the brain, including the hippocampus. Changes in hippocampal synaptic plasticity and subsequent impairment in cognitive functions such as learning and memory, are well documented in animal models of type 1 and type 2 diabetes. It is known that RAGE contributes to peripheral micro- and macro-vascular complications of diabetes. However, it is still unknown if RAGE plays a similar role in the development of CNS complications of diabetes. Therefore, we hypothesize that RAGE contributes to cognitive dysfunction, such as learning and memory impairments, in a mouse model of STZ-induced hyperglycemia. Control and STZ-induced hyperglycemic mice from WT and RAGE-KO groups were used for the behavioral experiments. While STZ-induced hyperglycemia decreased locomotor activity in the open field (OF) test, it did not affect the recognition memory in the novel object recognition (NOR) test in either genotype. Spatial memory, however, was impaired in STZ-induced hyperglycemic mice in WT but not in RAGE-KO group in both the Barnes maze (BM) and the Morris water maze (MWM) tests. Consistently, the RAGE antagonist FPS-ZM1 protected WT STZ-induced hyperglycemic mice from spatial memory impairment in the BM test. Our findings indicate that the parameters associated with locomotor activity and recognition memory were independent of RAGE in STZ-induced hyperglycemic mice. In contrast, the parameters associated with hippocampal-dependent spatial memory were dependent on RAGE expression.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Animals , Hippocampus , Hyperglycemia/complications , Maze Learning , Memory Disorders/etiology , Mice , Spatial Memory
5.
Front Cell Neurosci ; 14: 581075, 2020.
Article in English | MEDLINE | ID: mdl-33192327

ABSTRACT

It was long thought that astrocytes, given their lack of electrical signaling, were not involved in communication with neurons. However, we now know that one astrocyte on average maintains and regulates the extracellular neurotransmitter and potassium levels of more than 140,000 synapses, both excitatory and inhibitory, within their individual domains, and form a syncytium that can propagate calcium waves to affect distant cells via release of "gliotransmitters" such as glutamate, ATP, or adenosine. Neuromodulators can affect signal-to-noise and frequency transmission within cortical circuits by effects on inhibition, allowing for the filtering of relevant vs. irrelevant stimuli. Moreover, synchronized "resting" and desynchronized "activated" brain states are gated by short bursts of high-frequency neuromodulatory activity, highlighting the need for neuromodulation that is robust, rapid, and far-reaching. As many neuromodulators are released in a volume manner where degradation/uptake and the confines of the complex CNS limit diffusion distance, we ask the question-are astrocytes responsible for rapidly extending neuromodulator actions to every synapse? Neuromodulators are known to influence transitions between brain states, leading to control over plasticity, responses to salient stimuli, wakefulness, and sleep. These rapid and wide-spread state transitions demand that neuromodulators can simultaneously influence large and diverse regions in a manner that should be impossible given the limitations of simple diffusion. Intriguingly, astrocytes are ideally situated to amplify/extend neuromodulator effects over large populations of synapses given that each astrocyte can: (1) ensheath a large number of synapses; (2) release gliotransmitters (glutamate/ATP/adenosine) known to affect inhibition; (3) regulate extracellular potassium that can affect excitability and excitation/inhibition balance; and (4) express receptors for all neuromodulators. In this review article, we explore the hypothesis that astrocytes extend and amplify neuromodulatory influences on neuronal networks via alterations in calcium dynamics, the release of gliotransmitters, and potassium homeostasis. Given that neuromodulatory networks are at the core of our sleep-wake cycle and behavioral states, and determine how we interact with our environment, this review article highlights the importance of basic astrocyte function in homeostasis, general cognition, and psychiatric disorders.

6.
J Neurosci Res ; 98(5): 964-977, 2020 05.
Article in English | MEDLINE | ID: mdl-32067254

ABSTRACT

Changes in extracellular potassium ([K+ ]e ) modulate neuronal networks via changes in membrane potential, voltage-gated channel activity, and alteration to transmission at the synapse. Given the limited extracellular space in the central nervous system, potassium clearance is crucial. As activity-induced potassium transients are rapidly managed by astrocytic Kir4.1 and astrocyte-specific Na+ /K+ -ATPase, any neurotransmitter/neuromodulator that can regulate their function may have indirect influence on network activity. Neuromodulators differentially affect cortical/thalamic networks to align sensory processing with differing behavioral states. Given serotonin (5HT), norepinephrine (NE), and acetylcholine (ACh) differentially affect spike frequency adaptation and signal fidelity ("signal-to-noise") in somatosensory cortex, we hypothesize that [K+ ]e may be differentially regulated by the different neuromodulators to exert their individual effects on network function. This study aimed to compare effects of individually applied 5HT, NE, and ACh on regulating [K+ ]e in connection to effects on cortical-evoked response amplitude and adaptation in male mice. Using extracellular field and K+ ion-selective recordings of somatosensory stimulation, we found that differential effects of 5HT, NE, and ACh on [K+ ]e regulation mirrored differential effects on amplitude and adaptation. 5HT effects on transient K+ recovery, adaptation, and field post-synaptic potential amplitude were disrupted by barium (200 µM), whereas NE and ACh effects were disrupted by ouabain (1 µM) or iodoacetate (100 µM). Considering the impact [K+ ]e can have on many network functions; it seems highly efficient that neuromodulators regulate [K+ ]e to exert their many effects. This study provides functional significance for astrocyte-mediated buffering of [K+ ]e in neuromodulator-mediated shaping of cortical network activity.


Subject(s)
Acetylcholine/pharmacology , Astrocytes/drug effects , Membrane Potentials/drug effects , Norepinephrine/pharmacology , Potassium/metabolism , Serotonin/pharmacology , Somatosensory Cortex/drug effects , Animals , Astrocytes/metabolism , Glycolysis/physiology , Male , Mice , Signal Transduction/drug effects , Somatosensory Cortex/metabolism
7.
Neurochem Res ; 45(1): 100-108, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31254249

ABSTRACT

Astrocytes are highly dynamic cells that modulate synaptic transmission within a temporal domain of seconds to minutes in physiological contexts such as Long-Term Potentiation (LTP) and Heterosynaptic Depression (HSD). Recent studies have revealed that astrocytes also modulate a faster form of synaptic activity (milliseconds to seconds) known as Transient Heterosynaptic Depression (tHSD). However, the mechanism underlying astrocytic modulation of tHSD is not fully understood. Are the traditional gliotransmitters ATP or glutamate released via hemichannels/vesicles or are other, yet, unexplored pathways involved? Using various approaches to manipulate astrocytes, including the Krebs cycle inhibitor fluoroacetate, connexin 43/30 double knockout mice (hemichannels), and inositol triphosphate type-2 receptor knockout mice, we confirmed early reports demonstrating that astrocytes are critical for tHSD. We also confirmed the importance of group II metabotropic glutamate receptors (mGluRs) in astrocytic modulation of tHSD using a group II agonist. Using dominant negative SNARE mice, which have disrupted glial vesicle function, we also found that vesicular release of gliotransmitters and activation of adenosine A1 receptors are not required for tHSD. As astrocytes can release lipids upon receptor stimulation, we asked if astrocyte-derived endocannabinoids are involved in tHSD. Interestingly, a cannabinoid receptor 1 (CB1R) antagonist blocked and an inhibitor of the endogenous endocannabinoid 2-arachidonyl glycerol (2-AG) degradation potentiates tHSD in hippocampal slices. Taken together, this study provides the first evidence for group II mGluR-mediated astrocytic endocannabinoids in transiently suppressing presynaptic neurotransmitter release associated with the phenomenon of tHSD.


Subject(s)
Astrocytes/metabolism , Endocannabinoids/metabolism , Long-Term Synaptic Depression/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Astrocytes/drug effects , Endocannabinoids/antagonists & inhibitors , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Synaptic Depression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Piperidines/pharmacology , Pyrazoles/pharmacology , Synapses/drug effects , Synaptic Transmission/drug effects
8.
Neurodegener Dis ; 19(2): 60-77, 2019.
Article in English | MEDLINE | ID: mdl-31600762

ABSTRACT

North American incidence of Alzheimer's disease (AD) is expected to more than double over the coming generation. Although genetic factors surrounding the production and clearance of amyloid-ß and phosphorylated tau proteins are known to be responsible for a subset of early-onset AD cases, they do not explain the pathogenesis of the far more prevalent sporadic late-onset variant of the disease. It is thus likely that lifestyle and environmental factors contribute to neurodegenerative processes implicated in the pathogenesis of AD. Herein, we review evidence that (1) excess sucrose consumption induces AD-associated liver pathologies and brain insulin resistance, (2) chronic stress overdrives activity of locus coeruleus neurons, leading to loss of function (a common event in neurodegeneration), (3) high-sugar diets and stress promote the loss of neuroprotective sex hormones in men and women, and (4) Western dietary trends set the stage for a lithium-deficient state. We propose that these factors may intersect as part of a "perfect storm" to contribute to the widespread prevalence of neurodegeneration and AD. In addition, we put forth the argument that exercise and supplementation with trace lithium can counteract many of the deleterious consequences associated with excessive caloric intake and perpetual stress. We conclude that lifestyle and environmental factors likely contribute to AD pathogenesis and that simple lifestyle and dietary changes can help counteract their effects.


Subject(s)
Alzheimer Disease/etiology , Diet/adverse effects , Sedentary Behavior , Stress, Psychological/complications , Alzheimer Disease/pathology , Brain/pathology , Humans , Stress, Psychological/pathology
9.
Metab Brain Dis ; 33(5): 1563-1571, 2018 10.
Article in English | MEDLINE | ID: mdl-29862455

ABSTRACT

Alzheimer's disease (AD) is the sixth leading cause of death in the United States where it is estimated that one in three seniors dies with AD or another dementia. Are modern lifestyle habits a contributing factor? Increased carbohydrate (sugar) consumption, stress and disruption of sleep patterns are quickly becoming the norm rather than the exception. Interestingly, seven months on a non-invasive high sucrose diet (20% sucrose in drinking water) has been shown to induce behavioral, metabolic and pathological changes consistent with AD in wild-type mice. As chronic stress and depression are associated with loss of locus coeruleus (LC) noradrenergic neurons and projections (source of anti-inflammatory and trophic factor control), we assessed the ability for a selective LC neurotoxin (DSP4) to accelerate and aggravate a high-sucrose mediated AD-related phenotype in wild-type mice. Male C57/Bl6 mice were divided into four groups: 1) saline injected, 2) DSP4 injected, 3) high sucrose drinking water (20%) or 4) DSP4 injected and high sucrose drinking water. We demonstrate that high sucrose consumption and DSP4 treatment promote an early-stage AD-related phenotype after only 3-4 months, as evidenced by elevated fecal corticosterone, increased despair, spatial memory deficits, increased AChE activity, elevated NO production, decreased pGSK3ß and increased pTau. Combined treatment appears to accelerate and aggravate pathological processes consistent with Alzheimer disease and dementia. Developing a simple model in wild-type mice will highlight environmental and lifestyle factors that need to be addressed to slow, prevent or even reverse the rising trend in dementia patient numbers and cost.


Subject(s)
Alzheimer Disease/pathology , Behavior, Animal/drug effects , Benzylamines/pharmacology , Brain/drug effects , Dietary Sugars/administration & dosage , Neurotoxins/pharmacology , Sucrose/administration & dosage , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Diet , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Motor Activity/drug effects , Phosphorylation , tau Proteins/metabolism
10.
Mol Cell Neurosci ; 88: 53-61, 2018 04.
Article in English | MEDLINE | ID: mdl-29277734

ABSTRACT

In the somatosensory cortex, inhibitory networks are involved in low frequency sensory input adaptation/habituation that can be observed as a paired-pulse depression when using a dual stimulus electrophysiological paradigm. Given that astrocytes have been shown to regulate inhibitory interneuron activity, we hypothesized that astrocytes are involved in cortical sensory adaptation/habituation and constitute effectors of the 5HT-mediated increase in frequency transmission. Using extracellular recordings of evoked excitatory postsynaptic potentials (eEPSPs) in layer II/III of somatosensory cortex, we used various pharmacological approaches to assess the recruitment of astrocyte signaling in paired-pulse depression and serotonin-mediated increase in the paired-pulse ratio (pulse 2/pulse 1). In the absence of neuromodulators or pharmacological agents, the first eEPSP is much larger in amplitude than the second due to the recruitment of long-lasting evoked GABAA-dependent inhibitory activity from the first stimulus. Disruption of glycolysis or mGluR5 signaling resulted in a very similar loss of paired-pulse depression in field recordings. Interestingly, paired-pulse depression was similarly sensitive to disruption by ATP P2Y and adenosine A2A receptor antagonists. In addition, we show that pharmacological disruption of paired-pulse depression by mGluR5, P2Y, and glycolysis inhibition precluded serotonin effects on frequency transmission (typically increased the paired-pulse ratio). These data highlight the possibility for astrocyte involvement in cortical inhibitory activity seen in this simple cortical network and that serotonin may act on astrocytes to exert some aspects of its modulatory influence.


Subject(s)
Astrocytes/drug effects , Neurotransmitter Agents/pharmacology , Serotonin/pharmacology , Synaptic Transmission/drug effects , Animals , Electric Stimulation/methods , Hippocampus/drug effects , Interneurons/drug effects , Male , Membrane Potentials/drug effects , Mice, Inbred C57BL , Synaptic Transmission/physiology
11.
J Neuroinflammation ; 12: 37, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25890242

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS)-mediated sickness behaviour is known to be a result of increased inflammatory cytokines in the brain. Inflammatory cytokines have been shown to mediate increases in brain excitation by loss of GABAA-mediated inhibition through receptor internalization or inactivation. Inflammatory pathways, reactive oxygen species and stress are also known to increase monoamine oxidase-A (MAO-A) and acetylcholinesterase (ACh-E) activity. Given that neuromodulator actions on neural circuits largely depend on inhibitory pathways and are sensitive to alteration in corresponding catalytic enzyme activities, we assessed the impact of systemic LPS on neuromodulator-mediated shaping of a simple cortical network. METHODS: Extracellular field recordings of evoked postsynaptic potentials in adult mouse somatosensory cortical slices were used to evaluate effects of a single systemic LPS challenge on neuromodulator function 1 week later. Neuromodulators were administered transiently as a bolus (100 µl) to the bath perfusate immediately upstream of the recording site to mimic phasic release of neuromodulators and enable assessment of response temporal dynamics. RESULTS: Systemic LPS administration resulted in loss of both spontaneous and evoked inhibition as well as alterations in the temporal dynamics of neuromodulator effects on a paired-pulse paradigm. The effects on neuromodulator temporal dynamics were sensitive to the Monoamine oxidase-A (MAO-A) antagonist clorgyline (for norepinephrine and serotonin) and the ACh-E inhibitor donepezil (for acetylcholine). This is consistent with significant increases in total MAO and ACh-E activity found in hemi-brain samples from the LPS-treated group, supporting the notion that systemic LPS administration may lead to longer-lasting changes in inhibitory network function and enzyme (MAO/ACh-E) activity responsible for reduced neuromodulator actions. CONCLUSIONS: Given the significant role of neuromodulators in behavioural state and cognitive processes, it is possible that an inflammatory-mediated change in neuromodulator action plays a role in LPS-induced cognitive effects and could help define the link between infection and neuropsychiatric/degenerative conditions.


Subject(s)
Acetylcholinesterase/metabolism , Cerebral Cortex , Lipopolysaccharides/pharmacology , Monoamine Oxidase/metabolism , Neurons/drug effects , Acetylcholine/pharmacology , Action Potentials/drug effects , Animals , Bicuculline/pharmacology , Biogenic Monoamines/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , GABA-A Receptor Antagonists/pharmacology , Hindlimb Suspension/methods , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Reaction Time/drug effects , Signal Transduction/drug effects , Time Factors
12.
Biomed Res Int ; 2014: 810371, 2014.
Article in English | MEDLINE | ID: mdl-25147815

ABSTRACT

Matrix metalloproteinases (MMPs) significantly contribute to ischemia reperfusion (I/R) injury, namely, by the degradation of contractile proteins. However, due to the experimental models adopted and lack of isoform specificity of MMP inhibitors, the cellular source and identity of the MMP(s) involved in I/R injury remain to be elucidated. Using isolated adult rat cardiomyocytes, subjected to chemically induced I/R-like injury, we show that specific inhibition of MMP-2 expression and activity using MMP-2 siRNA significantly protected cardiomyocyte contractility from I/R-like injury. This was also associated with increased expression of myosin light chains 1 and 2 (MLC1/2) in comparison to scramble siRNA transfection. Moreover, the positive effect of MMP-2 siRNA transfection on cardiomyocyte contractility and MLC1/2 expression levels was also observed under control conditions, suggesting an important additional role for MMP-2 in physiological sarcomeric protein turnover. This study clearly demonstrates that intracellular expression of MMP-2 plays a significant role in sarcomeric protein turnover, such as MLC1 and MLC2, under aerobic (physiological) conditions. In addition, this study identifies intracellular/autocrine, cardiomyocyte-produced MMP-2, rather than paracrine/extracellular, as responsible for the degradation of MLC1/2 and consequent contractile dysfunction in cardiomyocytes subjected to I/R injury.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Muscle Contraction/physiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , RNA, Small Interfering/metabolism , Animals , Cardiac Myosins/metabolism , Male , Myosin Light Chains/metabolism , Protective Agents/metabolism , Rats , Rats, Sprague-Dawley
14.
Br J Pharmacol ; 170(2): 380-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23822644

ABSTRACT

BACKGROUND AND PURPOSE: Phosphorylation and degradation of myosin light chain 1 (MLC1) during myocardial ischaemia/reperfusion (I/R) injury is a well-established phenomenon. It has been established that MMP-2 is involved in MLC1 degradation and that this degradation is increased when MLC1 is phosphorylated. We hypothesized that simultaneous inhibition of MLC1 phosphorylation and MMP-2 activity will protect hearts from I/R injury. As phosphorylation of MLC1 and MMP-2 activity is important for normal heart function, we used a cocktail consisting combination of low (subthreshold for any protective effect alone) doses of MLC kinase, MMP-2 inhibitors and subthreshold dose of an MLC phosphatase activator. EXPERIMENTAL APPROACH: Isolated rat hearts were subjected to 20 min of global, no-flow ischaemia and 30 min reperfusion in the absence and presence of inhibitors of MLC1 phosphorylation and degradation. KEY RESULTS: The recovery of cardiac function was improved in a concentration-dependent manner by the MLC kinase inhibitor, ML-7 (1-5 µM), the MLC phosphatase activator, Y-27632 (0.05-1 µM) or the MMP inhibitor, doxycycline (Doxy, 1-30 µM). Co-administration of subthreshold doses of ML-7 (1 µM) and Y-27632 (0.05 µM) showed a potential synergistic effect in protecting cardiac contractility and MLC1 levels in I/R hearts. Further combination with a subthreshold concentration of Doxy (1 µM) showed additional protection that resulted in full recovery to control levels. CONCLUSIONS AND IMPLICATIONS: The results of this study exemplify a novel low-dose multidrug approach to pharmacological prevention of reperfusion injury that will enable a reduction of unwanted side effects and/or cytotoxicity associated with currently available MMP-2 and kinase inhibiting drugs.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Myocardial Reperfusion Injury/drug therapy , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Amides/administration & dosage , Amides/pharmacology , Animals , Azepines/administration & dosage , Azepines/pharmacokinetics , Azepines/pharmacology , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Doxycycline/administration & dosage , Doxycycline/pharmacology , Drug Synergism , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Male , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase Inhibitors/administration & dosage , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Reperfusion Injury/physiopathology , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/administration & dosage , Naphthalenes/pharmacokinetics , Naphthalenes/pharmacology , Phosphorylation/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
15.
J Cereb Blood Flow Metab ; 32(12): 2135-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872230

ABSTRACT

Given the brain's uniquely high cell density and tissue oxygen levels bordering on hypoxia, the ability to rapidly and precisely match blood flow to constantly changing patterns in neural activity is an essential feature of cerebrovascular regulation. Locus coeruleus-norepinephrine (LC-NE) projections innervate the cerebral vasculature and can mediate vasoconstriction. However, function of the LC-mediated constriction in blood-flow regulation has never been addressed. Here, using intrinsic optical imaging coupled with an anesthesia regimen that only minimally interferes with LC activity, we show that NE enhances spatial and temporal aspects of functional hyperemia in the mouse somatosensory cortex. Increasing NE levels in the cortex using an α(2)-adrenergic receptor antagonist paradoxically reduces the extent of functional hyperemia while enhancing the surround blood-flow reduction. However, the NE-mediated vasoconstriction optimizes spatial and temporal focusing of the hyperemic response resulting in a sixfold decrease in the disparity between blood volume and oxygen demand. In addition, NE-mediated vasoconstriction accelerated redistribution to subsequently active regions, enhancing temporal synchronization of blood delivery. These observations show an important role for NE in optimizing neurovascular coupling. As LC neuron loss is prominent in Alzheimer and Parkinson diseases, the diminished ability to couple blood volume to oxygen demand may contribute to their pathogenesis.


Subject(s)
Blood Volume , Cerebrovascular Circulation , Hyperemia , Locus Coeruleus , Norepinephrine/blood , Oxygen/blood , Somatosensory Cortex , Alzheimer Disease/blood , Alzheimer Disease/physiopathology , Animals , Blood Flow Velocity , Hyperemia/blood , Hyperemia/physiopathology , Locus Coeruleus/metabolism , Locus Coeruleus/physiopathology , Male , Mice , Parkinson Disease/blood , Parkinson Disease/physiopathology , Somatosensory Cortex/blood supply , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiopathology , Vasoconstriction
16.
Proc Natl Acad Sci U S A ; 108(2): 846-51, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187412

ABSTRACT

Aquaporin-4 (AQP4) is a primary influx route for water during brain edema formation. Here, we provide evidence that brain swelling triggers Ca(2+) signaling in astrocytes and that deletion of the Aqp4 gene markedly interferes with these events. Using in vivo two-photon imaging, we show that hypoosmotic stress (20% reduction in osmolarity) initiates astrocytic Ca(2+) spikes and that deletion of Aqp4 reduces these signals. The Ca(2+) signals are partly dependent on activation of P2 purinergic receptors, which was judged from the effects of appropriate antagonists applied to cortical slices. Supporting the involvement of purinergic signaling, osmotic stress was found to induce ATP release from cultured astrocytes in an AQP4-dependent manner. Our results suggest that AQP4 not only serves as an influx route for water but also is critical for initiating downstream signaling events that may affect and potentially exacerbate the pathological outcome in clinical conditions associated with brain edema.


Subject(s)
Aquaporin 4/chemistry , Aquaporin 4/genetics , Astrocytes/metabolism , Brain Edema/metabolism , Calcium/metabolism , Adenosine Triphosphate/chemistry , Animals , Brain/pathology , Edema/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmosis , Photons , Signal Transduction , Water/chemistry
17.
Cereb Cortex ; 18(12): 2789-95, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18372288

ABSTRACT

The locus coeruleus (LC) provides the sole source of norepinephrine (NE) to the cortex for modulation of cortical synaptic activity in response to salient sensory information. NE has been shown to improve signal-to-noise ratios, sharpen receptive fields and function in learning, memory, and cognitive performance. Although LC-mediated effects on neurons have been addressed, involvement of astrocytes has thus far not been demonstrated in these neuromodulatory functions. Here we show for the 1st time in live mice, that astrocytes exhibit rapid Ca(2+) increases in response to electrical stimulation of the LC. Additionally, robust peripheral stimulation known to result in phasic LC activity leads to Ca(2+) responses in astrocytes throughout sensory cortex that are independent of sensory-driven glutamate-dependent pathways. Furthermore, the astrocytic Ca(2+) transients are competitively modulated by alpha(2)-specific agonist/antagonist combinations known to impact LC output, are sensitive to the LC-specific neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, and are inhibited locally by an alpha-adrenergic antagonist. Future investigations of LC function must therefore consider the possibility that LC neuromodulatory effects are in part derived from activation of astrocytes.


Subject(s)
Astrocytes/physiology , Cerebral Cortex/physiology , Locus Coeruleus/physiology , Neurons/physiology , Receptors, Adrenergic, alpha/physiology , Animals , Calcium/physiology , Electric Stimulation , Isoflurane , Male , Mice , Mice, Inbred Strains , Receptors, Metabotropic Glutamate/physiology , Sensory Receptor Cells/physiology , Sensory Thresholds/physiology , Somatosensory Cortex/physiology
18.
Glia ; 50(4): 407-416, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15846795

ABSTRACT

Extracellular [K+] can range within 2.5-3.5 mM under normal conditions to 50-80 mM under ischemic and spreading depression events. Sustained exposure to elevated [K+]o has been shown to cause significant neuronal death even under conditions of abundant glucose supply. Astrocytes are well equipped to buffer this initial insult of elevated [K] through extensive gap junctional coupling, Na+/K+ pump activity (with associated glycogen and glycolytic potential), and endfoot siphoning capability. Their abundant energy availability and alkalinizing mechanisms help sustain Na+/K+ ATPase activity under ischemic conditions. Furthermore, passive K+ uptake mechanisms and water flux mediated through aquaporin-4 channels in endfoot processes are important energy-independent mechanisms. Unfortunately, as the length of ischemic episode is prolonged, these mechanisms increase to a point where they begin to have repercussions on other important cellular functions. Alkalinizing mechanisms induce an elevation of [Na+]i, increasing the energy demand of Na+/K+ ATPase and leading to eventual detrimental reversal of the Na+/glutamate- cotransporter and excitotoxic damage. Prolonged ischemia also results in cell swelling and activates volume regulatory processes that release excessive excitatory amino acids, further exacerbating excitotoxic injury. In the days following ischemic injury, reactive astrocytes demonstrate increased cell size and process thickness, leading to improved spatial buffering capacity in regions outside the lesion core where there is better neuronal survival. There is a substantial heterogeneity among reactive astrocytes, with some close to the lesion showing decreased buffering capacity. However, it appears that both Na+/K+ ATPase activity (along with energy production processes) as well as passive K+ uptake mechanisms are upregulated in gliotic tissue outside the lesion to enhance the above-mentioned homeostatic mechanisms.


Subject(s)
Brain Ischemia/metabolism , Homeostasis/physiology , Potassium/metabolism , Animals , Brain Ischemia/physiopathology , Humans
19.
J Neurophysiol ; 93(3): 1699-709, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15738276

ABSTRACT

Voltage-gated potassium channels are well established as critical for setting action potential frequency, membrane potential, and neurotransmitter release in neurons. However, their role in the "nonexcitable" glial cell type is yet to be fully understood. We used whole cell current kinetics, pharmacology, immunocytochemistry, and RT-PCR to characterize A-type current in hippocampal astrocyte cultures to better understand its function. Pharmacological analysis suggests that approximately 70, 10, and <5% of total A current is associated with Kv4, Kv3, and Kv1 channels, respectively. In addition, pharmacology and kinetics provide evidence for a significant contribution of KChIP accessory proteins to astrocytic A-channel composition. Localization of the Shaw Kv3.4 channel to astrocytic processes and the Shal Kv4.3 channel to soma suggest that these channels serve a specific function. Given this complex A-type channel expression pattern, we assessed the role of A currents in membrane voltage oscillations in response to current injections. Although TEA-sensitive delayed-rectifying currents are involved in the extent of repolarization, 4-AP-sensitive A currents serve to increase the rate. As in neurons, this effect may enable astrocytes to respond rapidly to high-frequency synaptic events. Our results indicate that hippocampal astrocytes in vitro express multiple A-type Kv channel alpha-subunits with accessory, possibly Ca(2+)-sensitive, cytoplasmic subunits that appear to be specifically localized to subcellular membrane compartments. Function of these channels remains to be determined in a physiological setting. However, this study suggests that they enable astrocytes to respond rapidly with membrane voltage oscillations to high-frequency incoming signals, possibly synchronizing astrocyte function to neuronal activity.


Subject(s)
Astrocytes/metabolism , Hippocampus/cytology , Potassium Channels, Voltage-Gated/metabolism , 4-Aminopyridine/pharmacology , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , Blotting, Northern/methods , Calcium/metabolism , Cells, Cultured , Electric Stimulation/methods , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Humans , Immunohistochemistry/methods , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Channel Gating/radiation effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/classification , Potassium Channels, Voltage-Gated/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Tetraethylammonium/pharmacology , Transfection/methods
20.
Brain Res ; 1024(1-2): 193-202, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15451382

ABSTRACT

Vimentin-expressing astrocytes in the adult are commonly associated with the proximal, most reactive gliotic response ultimately leading to the formation of a new glial limitans. It was thought, since vimentin expression and astroglial proliferation are most prominent nearest the lesion site, that vimentin may be a characteristic of immature newly divided astrocytes. We recently established a unique distribution of vimentin-expressing reactive astrocytes at the base of a focal cortical ischemic lesion in rats. The purpose of the present study was to assess the correlation of proliferation and migration with this unique distribution following focal injury. With the use of bromodeoxyuridine (BrdU) and immunohistochemistry for astrocytes and microglia/macrophages, proliferation and migration of cells was shown to be throughout the ipsilateral hemisphere on day one and become progressively more centralized to the lesion by day 3. The vimentin-expressing area at the base of the lesion does not exhibit distinguishable proliferation rates from non-vimentin-expressing regions surrounding the lesion and did not demonstrate obvious double labeling with BrdU+ cells, although on occasion vimentin expression is closely associated with BrdU. However, this region did become a focal point for migration into and around the lesion by day 3. Additionally, asymmetrical distribution of vimentin was shown in four different injury models with vimentin+ cells always situated between the lesion and the corpus callosum. It is concluded that although vimentin-expressing cells did not correlate with proliferating cells in these focal injury models, perhaps this distinct population of reactive astrocytes serve as a source of cytokines or as a physical conduit for migrating cells from distant sites through the corpus callosum.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Cell Movement/physiology , Cell Proliferation , Vimentin/biosynthesis , Animals , Astrocytes/cytology , Male , Rats , Rats, Wistar , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL