Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Eur J Pain ; 18(2): 223-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23857727

ABSTRACT

BACKGROUND: The function of brain networks can be changed in a maladaptive manner in response to chronic neuropathic pain. Analgesics can reduce pain by acting on such networks via direct or indirect (peripheral or spinal) mechanisms. This investigation aimed to map gabapentin's pharmacodynamics (PD) in the rodent brain following induction of neuropathic pain in order to further understand its PD profile. METHODS: Pharmacological magnetic resonance imaging (phMRI) and a novel functional connectivity analysis procedure were performed following vehicle or gabapentin treatment in the rat spinal nerve ligation (SNL) model of neuropathic pain as well as sham animals. RESULTS: phMRI performed in SNL animals revealed robust gabapentin-induced responses throughout the hippocampal formation, yet significant (p < 0.05, corrected for multiple comparisons) responses were also measured in other limbic structures and the sensorimotor system. In comparison, sham animals displayed weaker and less widespread phMRI signal changes subsequent to gabapentin treatment. Next, communities of networks possessing strong functional connectivity were elucidated in vehicle-treated SNL and sham animals. We observed that SNL and sham animals possessed distinct functional connectivity signatures. When measuring how gabapentin altered the behaviour of the discovered networks, a decrease in functional connectivity driven by gabapentin was not only observed, but the magnitude of this PD effect was greater in SNL animals. CONCLUSIONS: Using phMRI and functional connectivity analysis approaches, the PD effects of gabapentin in a preclinical neuropathic pain state were characterized. Furthermore, the current results offer insights on which brain systems gabapentin directly or indirectly acts upon.


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Neuralgia/drug therapy , Spinal Nerves/drug effects , gamma-Aminobutyric Acid/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Gabapentin , Male , Neuralgia/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Nerves/physiopathology
2.
Neuroscience ; 189: 299-304, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21619913

ABSTRACT

Latrepirdine is hypothesized to exert a unique mechanism of action involving stabilization of mitochondria that may have utility in treating Alzheimer's disease. However, the ability of latrepirdine to improve cognition in Alzheimer's disease (AD) is controversial due to a discrepancy between the positive signal reported in the multi-site phase II clinical trial where latrepirdine met all primary and secondary endpoints [Doody et al. (2008) Lancet 372:207-215], and the subsequent null effect observed in a multicenter, phase III trial. While dysfunction of mitochondria and abnormal energy metabolism has been linked to AD pathology, no studies have been reported that investigate latrepirdine's effect on cerebral glucose utilization (CGU). Glucose metabolism, following acute latrepirdine administration, can be used to help dose selection in Phase I dose-ranging studies. The aim of the current study was to assess changes in CGU in young and aged mice in vivo using [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) after acute treatment with latrepirdine. Two ages of B6SJLF2 mice (5 and 20 months old) were tested. Three test-retest FDG-PET baseline scans were assessed across all subjects. As CGU was heterogeneous in aged mice, compared to young mice, aged subjects were rank ordered and then counterbalanced into two CGU homogenous groups. In Studies 1 and 2, latrepirdine (1.0 mg/kg) significantly enhanced CGU in aged mice. In contrast, Study 3 revealed that latrepirdine did not modulate CGU in young mice. Monitoring changes in CGU in response to acute drug administration may represent an imaging biomarker for dose selection in AD. Further studies that would establish the translation from mice to non-human primates to humans need to be investigated to confirm the utility of FDG-PET in dose-selection for mitochondrial modulators.


Subject(s)
Aging/metabolism , Brain/drug effects , Fluorodeoxyglucose F18 , Glucose/metabolism , Indoles/pharmacology , Radiopharmaceuticals , Animals , Brain/diagnostic imaging , Brain/metabolism , Female , Mice , Positron-Emission Tomography
3.
Neuroscience ; 155(1): 263-9, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18534764

ABSTRACT

Neurovascular regulation, which is critical to the efficient functioning of the brain, is impaired in Alzheimer's disease and in transgenic mice overexpressing Abeta. Although senile plaques and neurofibrillary tangles represent neuropathological hallmarks of Alzheimer's disease, deposition of Abeta in cerebral blood vessels also likely plays a significant role in this debilitating and fatal disease. Further, soluble Abeta, which shows greater correlation with disease progression and severity than deposited plaques or tangles, displays strong vasoactive properties. The aim of this study was to develop a non-invasive model of cerebral vasoactivity that would ultimately be translatable to Alzheimer's disease as a marker for disease-modifying efficacy of novel small molecule and biologics drugs. Relative changes in cerebral blood volume following relevant doses of soluble Abeta(1-40) (0.01 or 0.1 mg/mouse), PBS, or the reverse peptide, Abeta(40-1) (0.01 or 0.1 mg/mouse), were monitored non-invasively by contrast-enhanced functional magnetic resonance imaging in anesthetized C57BL/6 mice. Experiments were performed on a 7T horizontal bore scanner using gradient echo echo-planar imaging. As expected, PBS and Abeta(40-1) did not induce any significant change in vascular response. In contrast, Abeta(1-40) significantly decreased CBV in a quantifiable, dose-related and region-specific manner. These data demonstrate for the first time the feasibility of characterizing pathogenic Abeta(1-40)-induced vascular dysfunction in vivo using a non-invasive approach. Further, this technique can be readily applied to preclinical screening in a longitudinal manner for novel drugs or antibodies targeting disease modification.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides , Brain/blood supply , Brain/pathology , Magnetic Resonance Imaging/methods , Peptide Fragments , Animals , Brain Mapping , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Image Processing, Computer-Assisted/methods , Mice , Oxygen/blood
5.
Br J Pharmacol ; 153(2): 367-79, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17965748

ABSTRACT

BACKGROUND AND PURPOSE: Activation of cannabinoid CB1 and/or CB2 receptors mediates analgesic effects across a broad spectrum of preclinical pain models. Selective activation of CB2 receptors may produce analgesia without the undesirable psychotropic side effects associated with modulation of CB1 receptors. To address selectivity in vivo, we describe non-invasive, non-ionizing, functional data that distinguish CB1 from CB2 receptor neural activity using pharmacological MRI (phMRI) in awake rats. EXPERIMENTAL APPROACH: Using a high field (7 T) MRI scanner, we examined and quantified the effects of non-selective CB1/CB2 (A-834735) and selective CB2 (AM1241) agonists on neural activity in awake rats. Pharmacological specificity was determined using selective CB1 (rimonabant) or CB2 (AM630) antagonists. Behavioural studies, plasma and brain exposures were used as benchmarks for activity in vivo. KEY RESULTS: The non-selective CB1/CB2 agonist produced a dose-related, region-specific activation of brain structures that agrees well with published autoradiographic CB1 receptor density binding maps. Pretreatment with a CB1 antagonist but not with a CB2 antagonist, abolished these activation patterns, suggesting an effect mediated by CB1 receptors alone. In contrast, no significant changes in brain activity were found with relevant doses of the CB2 selective agonist. CONCLUSION AND IMPLICATIONS: These results provide the first clear evidence for quantifying in vivo functional selectivity between CB1 and CB2 receptors using phMRI. Further, as the presence of CB2 receptors in the brain remains controversial, our data suggest that if CB2 receptors are expressed, they are not functional under normal physiological conditions.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Agonists , Algorithms , Animals , Behavior, Animal/drug effects , Cells, Cultured , Cerebrovascular Circulation/drug effects , Humans , Image Interpretation, Computer-Assisted , Inflammation/complications , Magnetic Resonance Imaging , Male , Motor Activity/drug effects , Pain/drug therapy , Pain/etiology , Peripheral Nervous System Diseases/complications , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors
7.
Br J Pharmacol ; 149(6): 761-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016515

ABSTRACT

BACKGROUND AND PURPOSE: To further assess the clinical potential of the blockade of metabotropic glutamate receptors (mGluR1) for the treatment of pain. EXPERIMENTAL APPROACH: We characterized the effects of A-841720, a novel, potent and non-competitive mGluR1 antagonist in models of pain and of motor and cognitive function. KEY RESULTS: At recombinant human and native rat mGluR1 receptors, A-841720 inhibited agonist-induced calcium mobilization, with IC50 values of 10.7+/-3.9 and 1.0 +/- 0.2 nM, respectively, while showing selectivity over other mGluR receptors, in addition to other neurotransmitter receptors, ion channels, and transporters. Intraperitoneal injection of A-841720 potently reduced complete Freund's adjuvant-induced inflammatory pain (ED50 = 23 micromol kg(-1)) and monoiodoacetate-induced joint pain (ED50 = 43 micromol kg(-1)). A-841720 also decreased mechanical allodynia observed in both the sciatic nerve chronic constriction injury and L5-L6 spinal nerve ligation (SNL) models of neuropathic pain (ED50 = 28 and 27 micromol kg(-1), respectively). Electrophysiological studies demonstrated that systemic administration of A-841720 in SNL animals significantly reduced evoked firing in spinal wide dynamic range neurons. Significant motor side effects were observed at analgesic doses and A-841720 also impaired cognitive function in the Y-maze and the Water Maze tests. CONCLUSIONS AND IMPLICATIONS: The analgesic effects of a selective mGluR1 receptor antagonist are associated with motor and cognitive side effects. The lack of separation between efficacy and side effects in pre-clinical models indicates that mGluR1 antagonism may not provide an adequate therapeutic window for the development of such antagonists as novel analgesic agents in humans.


Subject(s)
Analgesia , Cognition/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Motor Activity/drug effects , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Cells, Cultured , Fluorescence , Humans , Male , Rats , Rats, Sprague-Dawley
8.
Neuroscience ; 137(2): 583-91, 2006.
Article in English | MEDLINE | ID: mdl-16289887

ABSTRACT

Administration of ABT-594, a potent agonist for nicotinic acetylcholine receptors with selectivity for the alpha4beta2 receptor subtype, is known to modulate a diverse array of behaviors including those associated with nociception, anxiety and motor function. In this study, we sought to gain insight into the neural actions of ABT-594, in vivo, by conducting functional magnetic resonance imaging in awake and anesthetized rats. Using T(2)*-weighted gradient echo imaging and an ultrasmall superparamagnetic iron oxide contrast agent, functional imaging was conducted on a 4.7 T magnet to measure changes in relative cerebral blood volume. In awake, restrained, male Sprague-Dawley rats that were acclimated to the imaging environment, injection of ABT-594 (0.03-0.3 micromol/kg, i.v.) evoked changes to relative cerebral blood volume in several neural regions including the cingulate, somatosensory, motor, auditory, and pre-frontal cortices as well as the thalamus and the periaqueductal gray/dorsal raphe. These effects were typically bimodal with significant decreases in relative cerebral blood volume at the 0.03 micromol/kg dose and increases at the higher doses (0.1 and 0.3 micromol/kg). The decreases and increases in relative cerebral blood volume were often observed within the same region, but triggered by different doses. Both increases and decreases in relative cerebral blood volume were blocked by pretreatment with the noncompetitive nicotinic acetylcholine receptor antagonist, mecamylamine (5 micromol/kg, i.p.) in awake rats. Administration of ABT-594 (0.1 micromol/kg, i.v.) to alpha-chloralose-anesthetized rats did not significantly alter relative cerebral blood volume in any brain region suggesting an anesthetic-related interference with the effects of ABT-594. The neural regions affected by administration of ABT-594 corresponded well to the known pre-clinical behavioral profile for this compound, and demonstrate the utility of using functional magnetic resonance imaging in awake animals to study pharmacological action.


Subject(s)
Acetylcholine/metabolism , Azetidines/pharmacology , Brain/drug effects , Brain/metabolism , Cerebrovascular Circulation/drug effects , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Anesthetics, Intravenous/pharmacology , Animals , Brain/blood supply , Brain Mapping , Cerebrovascular Circulation/physiology , Chloralose/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Interactions/physiology , Magnetic Resonance Imaging/methods , Male , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Pain/metabolism , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Wakefulness/drug effects , Wakefulness/physiology
16.
J Neurochem ; 78(4): 704-14, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520891

ABSTRACT

2-N-Pentyl-4-pentynoic acid [pentyl-4-yn-valproic acid (VPA)] is an analogue of valproic acid that induces neuritogenesis and increases neural cell adhesion molecule (NCAM) prevalence in cultured neural cells. As memory consolidation involves synapse growth, aided by cell adhesion molecule function, we determined whether or not pentyl-4-yn-VPA had cognition-enhancing properties. Pentyl-4-yn-VPA (16-85 mg/kg) significantly improved water maze learning and task retention when given prior to each training session. Acute administration of pentyl-4-yn-VPA also influenced memory consolidation processes as, when given at 3 h post-passive avoidance training, the amnesia induced by scopolamine given 6 h post-training was prevented in a dose-dependent manner. Chronic administration of pentyl-4-yn-VPA (16.8 or 50.4 mg/kg) also significantly reduced escape latencies in the water maze task, 24 h following the last drug administration. This improved spatial learning was accompanied by enhanced neuroplasticity as the expression of NCAM polysialylated neurons in the infragranular zone of the dentate gyrus and in layer II of the perirhinal and piriform cortex was increased significantly following chronic drug treatment. The cognition-enhancing qualities of pentyl-4-yn-VPA, combined with its ability to attenuate the age-related loss of the NCAM polysialylation state, suggest that it may effectively slow the onset of cognitive decline.


Subject(s)
Avoidance Learning/physiology , Maze Learning/physiology , Neural Cell Adhesion Molecules/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Temporal Lobe/drug effects , Valproic Acid/pharmacology , Aging , Animals , Avoidance Learning/drug effects , Blood Pressure/physiology , Cell Line , Heart Rate/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Kidney/anatomy & histology , Kidney/drug effects , Liver/anatomy & histology , Liver/drug effects , Lung/anatomy & histology , Lung/drug effects , Male , Maze Learning/drug effects , Muscarinic Antagonists/pharmacology , Neural Cell Adhesion Molecules/chemistry , Neurons/metabolism , Protein Binding , Radioligand Assay , Rats , Rats, Wistar , Scopolamine/pharmacology , Temporal Lobe/metabolism , Valproic Acid/analogs & derivatives
17.
Eur J Pharmacol ; 424(1): 37-44, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11470258

ABSTRACT

NNC-711 [1-(2-((diphenylmethylene)amino)oxy)ethyl)-1,2,4,6-tetrahydro-3-pyridinecarboxylic acid hydrochloride], a gamma-aminobutyric acid (GABA) reuptake inhibitor with anticonvulsant activity, was investigated with respect to its cognition-enhancing and neuroprotective potency. In the rat, administration of NNC-711 immediately prior to training prevented amnesia for a passive avoidance task induced by the acetylcholine receptor antagonist scopolamine. NNC-711 was also effective in protecting against ischemia-induced death of CA1 pyramidal neurons in a model of bilateral common carotid artery occlusion in the gerbil. In addition to a neuroprotective activity, NNC-711 exhibited significant cognition-enhancing actions. Daily administration of NNC-711, immediately prior to a spatial learning task, significantly reduced escape latencies in the water maze paradigm in both mature (postnatal day 80) and aged (28 months) rats. All of the above actions exhibited a bell-shaped response with an optimal dose of 0.5-1.0 mg/kg. These investigations with NNC-711 and previous clinical observations on the structurally related anticonvulsant tiagabine confirm the potential of GABA reuptake inhibitors as anti-amnesia and cognition-enhancing agents.


Subject(s)
Cognition/drug effects , GABA Antagonists/pharmacology , Ischemic Attack, Transient/prevention & control , Nipecotic Acids/pharmacology , Oximes/pharmacology , Amnesia/chemically induced , Amnesia/prevention & control , Animals , Avoidance Learning/drug effects , Cell Survival/drug effects , Cognition/physiology , Dose-Response Relationship, Drug , Gerbillinae , Hippocampus/cytology , Hippocampus/drug effects , Ischemic Attack, Transient/physiopathology , Male , Maze Learning/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Scopolamine/administration & dosage
18.
J Neuropathol Exp Neurol ; 60(2): 132-40, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11273001

ABSTRACT

To investigate the role of polysialylated neural cell adhesion molecule (NCAM PSA)-mediated plasticity after injury, we examined the temporal and spatial expression of NCAM PSA immunoreactivity in the medial temporal lobe following global ischemia. Male Mongolian gerbils were subjected to bilateral common carotid artery occlusion for 5 min and killed at increasing times post-occlusion. The well-characterized delayed CAl pyramidal cell death was observed 5-7 days post-occlusion. At post-occlusion days 1-2 there was a small but significant increase of NCAM PSA-positive hippocampal granule cells followed by an equally significant decrease at post-occlusion day 5. In contrast, a substantial increase in glial PSA expression was observed in all hippocampal regions at 1-7 days post-occlusion that was associated generally with stellate astroglia and specifically with the radial processes of glia traversing the granule cell layer of the dentate gyrus. Administration of the glutamate antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-ben-zo(F)quinoxaline significantly blocked the ischemia-induced modulation of neuronal and glial NCAM PSA expression. Astroglial NCAM polysialylation became attenuated by 35 days post-occlusion except in the CAI area of cell death. The temporal and regional pattern of polysialylated NCAM expression in the ischemic gerbil hippocampus implicates this neuroplastic marker in mechanisms of neurotrophic-dependent repair/remodeling that ensue following transient interruption of blood flow.


Subject(s)
Hippocampus/pathology , Ischemic Attack, Transient/pathology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Neuroglia/pathology , Neurons/pathology , Sialic Acids/metabolism , Animals , Cell Count , Cell Death , Disease Models, Animal , Gerbillinae , Male , Pyramidal Cells/pathology
19.
J Neurobiol ; 45(3): 135-41, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11074459

ABSTRACT

Within the rat medial temporal lobe, transient modulations of neural cell adhesion molecule (NCAM) polysialylation have been observed to follow spatial learning. These have been attributed to neuroplastic events associated with the processing of information destined for long term memory consolidation. To determine if similar events are associated with avoidance learning, we investigated change in polysialylated cell number in the entorhinal, perirhinal, and piriform cortex, following acquisition of a passive avoidance task in the rat. Direct quantification of polysialylated neurons in layer II of these cortical regions revealed a significant increase in polysialylated cell frequency at 12 h following passive avoidance training. Unlike spatial learning, the increased expression of polysialylated neurons persisted for up to 24-48 h following training. In the more dorsal aspect of the perirhinal/entorhinal cortex, this increase was found to be specific to learning, as it was not observed in animals rendered amnesic with scopolamine. By contrast, change in polysialylated cell frequency in the ventral aspect of the medial temporal lobe was only partially reduced by amnesic doses of scopolamine. The persisting activation of NCAM polysialylation in the more dorsal aspects of the perirhinal and entorhinal cortex is suggested to reflect the need for more extensive synaptic alterations, as compared to those required for the consolidation of spatial learning. Moreover, the neuroplastic modulations observed in the more ventral regions of the entorhinal and perirhinal cortex appear to be a unique aspect of avoidance conditioning that reflects the activation of alternative learning strategies associated with motivational and/or contextual parameters of the task.


Subject(s)
Avoidance Learning/physiology , Cell Count/statistics & numerical data , Entorhinal Cortex/physiology , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Olfactory Pathways/physiology , Sialic Acids/metabolism , Animals , Avoidance Learning/drug effects , Biomarkers/analysis , Dendrites/metabolism , Dendrites/ultrastructure , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Male , Neural Pathways/cytology , Neurons/cytology , Neurons/drug effects , Olfactory Pathways/cytology , Olfactory Pathways/drug effects , Rats , Rats, Wistar , Scopolamine/pharmacology
20.
Am J Physiol ; 277(4): R1196-204, 1999 10.
Article in English | MEDLINE | ID: mdl-10516262

ABSTRACT

Thyrotropin-releasing hormone (TRH) and certain TRH analogs show substantial neuroprotective effects in experimental brain or spinal cord trauma but also have other physiological actions (autonomic, analeptic, and endocrine) that may be undesirable for the treatment of neurotrauma in humans. We developed a novel TRH analog (2-ARA-53a), with substitutions at the NH(2)-terminus and imidazole ring, that preserves the neuroprotective action of TRH-like compounds while decreasing or eliminating their autonomic, analeptic, and endocrine effects. Rats administered 2-ARA-53a (1.0 mg/kg, n = 17) intravenously 30 min after lateral fluid percussion brain injury showed marked improvement in motor recovery compared with vehicle-treated controls (n = 14). Treatment of mice subjected to moderate controlled cortical impact brain injury, at the same dose and time after trauma (n = 8), improved both motor recovery and cognitive performance in a water maze place learning task compared with vehicle-treated controls (n = 8). In injured rats, no autonomic or analeptic effects were observed with this compound, and endocrine effects were significantly reduced with 2-ARA-53a, in contrast to those found with a typical NH(2)-terminal-substituted TRH analog (YM-14673). These findings demonstrate that the neuroprotective effects of TRH-related compounds can be dissociated from their other major physiological actions and suggest a potential role for dual-substituted TRH analogs in the treatment of clinical neurotrauma.


Subject(s)
Brain Injuries/physiopathology , Brain Injuries/psychology , Cognition/drug effects , Motor Activity/drug effects , Thyrotropin-Releasing Hormone/analogs & derivatives , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiopathology , Azetidines/pharmacology , Brain Injuries/mortality , Central Nervous System Stimulants/pharmacology , Dipeptides/pharmacology , Endocrine Glands/drug effects , Endocrine Glands/physiopathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Nervous System/drug effects , Nervous System/physiopathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...