Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 37(5): 109950, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731619

ABSTRACT

Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Excitatory Amino Acid Agonists/pharmacology , Glycine/analogs & derivatives , Interneurons/drug effects , Prefrontal Cortex/drug effects , Receptors, Metabotropic Glutamate/agonists , Resorcinols/pharmacology , Schizophrenia/drug therapy , Schizophrenic Psychology , Somatostatin/metabolism , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Female , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Glycine/pharmacology , Interneurons/metabolism , Male , Memory, Short-Term/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/metabolism , Schizophrenia/physiopathology , Somatostatin/genetics
2.
Neuroimage Clin ; 30: 102633, 2021.
Article in English | MEDLINE | ID: mdl-33780866

ABSTRACT

PURPOSE: The sensitivity and accuracy of chemical exchange saturation transfer (CEST) and nuclear Overhauser enhancement (NOE) effects for assessing injury-associated changes in cervical spinal cords were evaluated in squirrel monkeys. Multiple interacting pools of protons, including one identified by an NOE at -1.6 ppm relative to water (NOE(-1.6)), were derived and quantified from fitting proton Z-spectra. The effects of down-sampled data acquisitions and corrections for non-specific factors including T1, semi-solid magnetization transfer, and direct saturation of free water (DS), were investigated. The overall goal is to develop a protocol for rapid data acquisition for assessing the molecular signatures of the injured spinal cord and its surrounding regions. METHODS: MRI scans were recorded of anesthetized squirrel monkeys at 9.4 T, before and after a unilateral dorsal column sectioning of the cervical spinal cord. Z-spectral images at 51 different RF offsets were acquired. The amplitudes of CEST and NOE effects from multiple proton pools were quantified using a six-pool Lorenzian fitting of each Z-spectrum (MTRmfit). In addition, down-sampled data using reduced selections of RF offsets were analyzed and compared. An apparent exchange-dependent relaxation (AREXmfit) method was also used to correct for non-specific factors in quantifying regional spectra around lesion sites. RESULTS: The parametric maps from multi-pool fitting using the complete sampling data (P51e) detected unilateral changes at and around the injury. The maps derived from selected twofold down-sampled data with appropriate interpolation (P26sI51) revealed quite similar spatial distributions of different pools as those obtained using P51e at each resonance shift. Across 10 subjects, both data acquisition schemes detected significant decreases in NOE(-3.5) and NOE(-1.6) and increases in DS(0.0) and CEST(3.5) at the lesion site relative to measures of the normal tissues before injury. AREXmfit of cysts and other abnormal tissues at and around the lesion site also exhibited significant changes, especially at 3.5, -1.6 and -3.5 ppm RF offsets. CONCLUSION: These results confirm that a reduced set of RF offsets and down sampling are adequate for CEST imaging of injured spinal cord and allow shorter imaging times and/or permit additional signal averaging. AREXmfit correction improved the accuracy of CEST and NOE measures. The results provide a rapid (~13 mins), sensitive, and accurate protocol for deriving multiple NOE and CEST effects simultaneously in spinal cord imaging at high field.


Subject(s)
Cervical Cord , Image Interpretation, Computer-Assisted , Algorithms , Cervical Cord/diagnostic imaging , Magnetic Resonance Imaging , Protons , Sensitivity and Specificity
3.
Brain Stimul ; 14(2): 261-272, 2021.
Article in English | MEDLINE | ID: mdl-33460838

ABSTRACT

Transcranial focused ultrasound (FUS) stimulation under MRI guidance, coupled with functional MRI (fMRI) monitoring of effects, offers a precise, noninvasive technology to dissect functional brain circuits and to modulate altered brain functional networks in neurological and psychiatric disorders. Here we show that ultrasound at moderate intensities modulated neural activity bi-directionally. Concurrent sonication of somatosensory areas 3a/3b with 250 kHz FUS suppressed the fMRI signals produced there by peripheral tactile stimulation, while at the same time eliciting fMRI activation at inter-connected, off-target brain regions. Direct FUS stimulation of the cortex resulted in different degrees of BOLD signal changes across all five off-target regions, indicating that its modulatory effects on active and resting neurons differed. This is the first demonstration of the dual suppressive and excitative modulations of FUS on a specific functional circuit and of ability of concurrent FUS and MRI to evaluate causal interactions between functional circuits with neuron-class selectivity.


Subject(s)
Brain , Magnetic Resonance Imaging , Animals , Brain/diagnostic imaging , Brain Mapping , Female , Humans , Primates , Rest
4.
NMR Biomed ; 33(4): e4216, 2020 04.
Article in English | MEDLINE | ID: mdl-31943383

ABSTRACT

Spinal cord injuries (SCIs) are a leading cause of disability and can severely impact the quality of life. However, to date, the processes of spontaneous repair of damaged spinal cord remain incompletely understood, partly due to a lack of appropriate longitudinal tracking methods. Noninvasive, multiparametric magnetic resonance imaging (MRI) provides potential biomarkers for the comprehensive evaluation of spontaneous repair after SCI. In this study in rats, a clinically relevant contusion injury was introduced at the lumbar level that impairs both hindlimb motor and sensory functions. Quantitative MRI measurements were acquired at baseline and serially post-SCI for up to 2 wk. The progressions of injury and spontaneous recovery in both white and gray matter were tracked longitudinally using pool-size ratio (PSR) measurements derived from quantitative magnetization transfer (qMT) methods, measurements of water diffusion parameters using diffusion tensor imaging (DTI) and intrasegment functional connectivity derived from resting state functional MRI. Changes in these quantitative imaging measurements were correlated with behavioral readouts. We found (a) a progressive decrease in PSR values within 2 wk post-SCI, indicating a progressive demyelination at the center of the injury that was validated with histological staining, (b) PSR correlated closely with fractional anisotropy and transverse relaxation of free water, but did not show significant correlations with behavioral recovery, and (c) preliminary evidence that SCI induced a decrease in functional connectivity between dorsal horns below the injury site at 24 h. Findings from this study not only confirm the value of qMT and DTI methods for assessing the myelination state of injured spinal cord but indicate that they may also have further implications on whether therapies targeted towards remyelination may be appropriate. Additionally, a better understanding of changes after SCI provides valuable information to guide and assess interventions.


Subject(s)
Behavior, Animal , Diffusion Tensor Imaging , Magnetic Resonance Imaging , Recovery of Function , Spinal Cord Injuries/diagnostic imaging , Spinal Cord Injuries/physiopathology , Animals , Anisotropy , Male , Rats, Sprague-Dawley , Reproducibility of Results , Spinal Cord/pathology , Spinal Cord/physiopathology
5.
PLoS One ; 13(4): e0191299, 2018.
Article in English | MEDLINE | ID: mdl-29698491

ABSTRACT

OBJECTIVE: We have previously demonstrated that insulin signaling, through the downstream signaling kinase Akt, is a potent modulator of dopamine transporter (DAT) activity, which fine-tunes dopamine (DA) signaling at the synapse. This suggests a mechanism by which impaired neuronal insulin receptor signaling, a hallmark of diet-induced obesity, may contribute to impaired DA transmission. We tested whether a short-term (two-week) obesogenic high-fat (HF) diet could reduce striatal Akt activity, a marker of central insulin, receptor signaling and blunt striatal and dopaminergic network responsiveness to amphetamine (AMPH). METHODS: We examined the effects of a two-week HF diet on striatal DAT activity in rats, using AMPH as a probe in a functional magnetic resonance imaging (fMRI) assay, and mapped the disruption in AMPH-evoked functional connectivity between key dopaminergic targets and their projection areas using correlation and permutation analyses. We used phosphorylation of the Akt substrate GSK3α in striatal extracts as a measure of insulin receptor signaling. Finally, we confirmed the impact of HF diet on striatal DA D2 receptor (D2R) availability using [18F]fallypride positron emission tomography (PET). RESULTS: We found that rats fed a HF diet for only two weeks have reductions in striatal Akt activity, a marker of decreased striatal insulin receptor signaling and blunted striatal responsiveness to AMPH. HF feeding also reduced interactions between elements of the mesolimbic (nucleus accumbens-anterior cingulate) and sensorimotor circuits (caudate/putamen-thalamus-sensorimotor cortex) implicated in hedonic feeding. D2R availability was reduced in HF-fed animals. CONCLUSION: These studies support the hypothesis that central insulin signaling and dopaminergic neurotransmission are already altered after short-term HF feeding. Because AMPH induces DA efflux and brain activation, in large part via DAT, these findings suggest that blunted central nervous system insulin receptor signaling through a HF diet can impair DA homeostasis, thereby disrupting cognitive and reward circuitry involved in the regulation of hedonic feeding.


Subject(s)
Brain/drug effects , Brain/metabolism , Diet, High-Fat/adverse effects , Dopamine/metabolism , Obesity/chemically induced , Obesity/metabolism , Amphetamine/pharmacology , Animals , Brain/pathology , Insulin/metabolism , Male , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Nerve Net/drug effects , Obesity/pathology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Time Factors
6.
Brain Imaging Behav ; 12(1): 87-95, 2018 02.
Article in English | MEDLINE | ID: mdl-28108946

ABSTRACT

Adjuvant chemotherapy has been used for decades to treat cancer, and it is well known that disruptions in cognitive function and memory are common chemotherapeutic adverse effects. However, studies using neuropsychological metrics have also reported group differences in cognitive function and memory before or without chemotherapy, suggesting that complex factors obscure the true etiology of chemotherapy-induced cognitive dysfunction (CICD) in humans. Therefore, to better understand possible mechanisms of CICD, we explored the effects of CICD in rats through cognition testing using novel object recognition (NOR) and contextual fear conditioning (CFC), and through metabolic neuroimaging via [18F]fluorodeoxyglucose (FDG) positron emission tomography (PET). Cancer-naïve, female Sprague-Dawley rats were administered either saline (1 mL/kg) or doxorubicin (DOX) (1 mg/kg in a volume of 1 mL/kg) weekly for five weeks (total dose = 5 mg/kg), and underwent cognition testing and PET imaging immediately following the treatment regime and 30 days post treatment. We did not observe significant differences with CFC testing post-treatment for either group. However, the chemotherapy group exhibited significantly decreased performance in the NOR test and decreased 18F-FDG uptake only in the prefrontal cortex 30 days post-treatment. These results suggest that long-term impairment within the prefrontal cortex is a plausible mechanism of CICD in this study, suggesting DOX-induced toxicity in the prefrontal cortex at the dose used.


Subject(s)
Antineoplastic Agents/toxicity , Brain/drug effects , Brain/diagnostic imaging , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/diagnostic imaging , Doxorubicin/toxicity , Animals , Brain Mapping , Cognitive Dysfunction/psychology , Conditioning, Psychological/drug effects , Disease Models, Animal , Fear/drug effects , Female , Fluorodeoxyglucose F18 , Magnetic Resonance Imaging , Neuroimaging , Positron-Emission Tomography , Radiopharmaceuticals , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Tomography, X-Ray Computed
7.
Magn Reson Med ; 79(5): 2773-2783, 2018 05.
Article in English | MEDLINE | ID: mdl-28905408

ABSTRACT

PURPOSE: Numerous studies have adopted resting-state functional MRI methods to infer functional connectivity between cortical regions, but very few have translated them to the spinal cord, despite its critical role in the central nervous system. Resting-state functional connectivity between gray matter horns of the spinal cord has previously been shown to be detectable in humans and nonhuman primates, but it has not been reported previously in rodents. METHODS: Resting-state functional MRI of the cervical spinal cord of live anesthetized rats was performed at 9.4 T. The quality of the functional images acquired was assessed, and quantitative analyses of functional connectivity in C4-C7 of the spinal cord were derived. RESULTS: Robust gray matter horn-to-horn connectivity patterns were found that were statistically significant when compared with adjacent control regions. Specifically, dorsal-dorsal and ventral-ventral connectivity measurements were most prominent, while ipsilateral dorsal-ventral connectivity was also observed but to a lesser extent. Quantitative evaluation of reproducibility also revealed moderate robustness in the bilateral sensory and motor networks that was weaker in the dorsal-ventral connections. CONCLUSIONS: This study reports the first evidence of resting-state functional circuits within gray matter in the rat spinal cord, and verifies their detectability using resting-state functional MRI at 9.4 T. Magn Reson Med 79:2773-2783, 2018. © 2017 International Society for Magnetic Resonance in Medicine.


Subject(s)
Cervical Cord/diagnostic imaging , Cervical Cord/physiology , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Signal Processing, Computer-Assisted , Animals , Gray Matter/diagnostic imaging , Gray Matter/physiology , Male , Rats , Rats, Sprague-Dawley
8.
Neuron ; 96(6): 1358-1372.e4, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29268098

ABSTRACT

Cholinergic regulation of dopaminergic inputs into the striatum is critical for normal basal ganglia (BG) function. This regulation of BG function is thought to be primarily mediated by acetylcholine released from cholinergic interneurons (ChIs) acting locally in the striatum. We now report a combination of pharmacological, electrophysiological, optogenetic, chemogenetic, and functional magnetic resonance imaging studies suggesting extra-striatal cholinergic projections from the pedunculopontine nucleus to the substantia nigra pars reticulata (SNr) act on muscarinic acetylcholine receptor subtype 4 (M4) to oppose cAMP-dependent dopamine receptor subtype 1 (D1) signaling in presynaptic terminals of direct pathway striatal spiny projections neurons. This induces a tonic inhibition of transmission at direct pathway synapses and D1-mediated activation of motor activity. These studies provide important new insights into the unique role of M4 in regulating BG function and challenge the prevailing hypothesis of the centrality of striatal ChIs in opposing dopamine regulation of BG output.


Subject(s)
Basal Ganglia/cytology , Cholinergic Neurons/physiology , Dopamine/metabolism , Pars Reticulata/physiology , Receptor, Muscarinic M4/metabolism , Acetylcholine/metabolism , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/physiology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/pharmacology , Cholinergic Neurons/drug effects , Dopamine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Locomotion/drug effects , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurotransmitter Agents/pharmacology , Oxygen/blood , Pars Reticulata/cytology , Pars Reticulata/diagnostic imaging , Pedunculopontine Tegmental Nucleus/cytology , Receptor, Muscarinic M4/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Neuron ; 86(4): 1029-1040, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25937172

ABSTRACT

Schizophrenia is associated with disruptions in N-methyl-D-aspartate glutamate receptor subtype (NMDAR)-mediated excitatory synaptic signaling. The metabotropic glutamate receptor subtype 5 (mGlu5) is a closely associated signaling partner with NMDARs and regulates NMDAR function in forebrain regions implicated in the pathology of schizophrenia. Efficacy of mGlu5 positive allosteric modulators (PAMs) in animal models of psychosis and cognition was previously attributed to potentiation of NMDAR function. To directly test this hypothesis, we identified VU0409551 as a novel mGlu5 PAM that exhibits distinct stimulus bias and selectively potentiates mGlu5 coupling to Gαq-mediated signaling but not mGlu5 modulation of NMDAR currents or NMDAR-dependent synaptic plasticity in the rat hippocampus. Interestingly, VU0409551 produced robust antipsychotic-like and cognition-enhancing activity in animal models. These data provide surprising new mechanistic insights into the actions of mGlu5 PAMs and suggest that modulation of NMDAR currents is not critical for in vivo efficacy. VIDEO ABSTRACT.


Subject(s)
Antipsychotic Agents/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Allosteric Regulation/drug effects , Animals , Cognition/drug effects , Cognition/physiology , Glutamic Acid/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Signal Transduction/drug effects
10.
Neuropsychopharmacology ; 39(7): 1578-93, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24442096

ABSTRACT

Accumulating evidence suggests that selective M4 muscarinic acetylcholine receptor (mAChR) activators may offer a novel strategy for the treatment of psychosis. However, previous efforts to develop selective M4 activators were unsuccessful because of the lack of M4 mAChR subtype specificity and off-target muscarinic adverse effects. We recently developed VU0152100, a highly selective M4 positive allosteric modulator (PAM) that exerts central effects after systemic administration. We now report that VU0152100 dose-dependently reverses amphetamine-induced hyperlocomotion in rats and wild-type mice, but not in M4 KO mice. VU0152100 also blocks amphetamine-induced disruption of the acquisition of contextual fear conditioning and prepulse inhibition of the acoustic startle reflex. These effects were observed at doses that do not produce catalepsy or peripheral adverse effects associated with non-selective mAChR agonists. To further understand the effects of selective potentiation of M4 on region-specific brain activation, VU0152100 alone and in combination with amphetamine were evaluated using pharmacologic magnetic resonance imaging (phMRI). Key neural substrates of M4-mediated modulation of the amphetamine response included the nucleus accumbens (NAS), caudate-putamen (CP), hippocampus, and medial thalamus. Functional connectivity analysis of phMRI data, specifically assessing correlations in activation between regions, revealed several brain networks involved in the M4 modulation of amphetamine-induced brain activation, including the NAS and retrosplenial cortex with motor cortex, hippocampus, and medial thalamus. Using in vivo microdialysis, we found that VU0152100 reversed amphetamine-induced increases in extracellular dopamine levels in NAS and CP. The present data are consistent with an antipsychotic drug-like profile of activity for VU0152100. Taken together, these data support the development of selective M4 PAMs as a new approach to the treatment of psychosis and cognitive impairments associated with psychiatric disorders such as schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Pyridines/pharmacology , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/chemistry , Thiophenes/pharmacology , Amphetamine/toxicity , Animals , Blood Pressure/drug effects , Brain/drug effects , Brain/pathology , Cell Line, Transformed , Central Nervous System Stimulants/toxicity , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Fear/drug effects , Heart Rate/drug effects , Humans , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M4/deficiency , Receptor, Muscarinic M4/genetics , Reflex, Startle/drug effects
11.
J Neurosci ; 32(25): 8532-44, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22723693

ABSTRACT

M(1) muscarinic acetylcholine receptors (mAChRs) represent a viable target for treatment of multiple disorders of the central nervous system (CNS) including Alzheimer's disease and schizophrenia. The recent discovery of highly selective allosteric agonists of M(1) receptors has provided a major breakthrough in developing a viable approach for the discovery of novel therapeutic agents that target these receptors. Here we describe the characterization of two novel M(1) allosteric agonists, VU0357017 and VU0364572, that display profound differences in their efficacy in activating M(1) coupling to different signaling pathways including Ca(2+) and ß-arrestin responses. Interestingly, the ability of these agents to differentially activate coupling of M(1) to specific signaling pathways leads to selective actions on some but not all M(1)-mediated responses in brain circuits. These novel M(1) allosteric agonists induced robust electrophysiological effects in rat hippocampal slices, but showed lower efficacy in striatum and no measureable effects on M(1)-mediated responses in medial prefrontal cortical pyramidal cells in mice. Consistent with these actions, both M(1) agonists enhanced acquisition of hippocampal-dependent cognitive function but did not reverse amphetamine-induced hyperlocomotion in rats. Together, these data reveal that M(1) allosteric agonists can differentially regulate coupling of M(1) to different signaling pathways, and this can dramatically alter the actions of these compounds on specific brain circuits important for learning and memory and psychosis.


Subject(s)
Behavior, Animal/drug effects , Benzamides/pharmacology , Biphenyl Compounds/pharmacology , Brain/drug effects , Muscarinic Agonists/pharmacology , Receptor, Muscarinic M1/agonists , Animals , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Cell Line , Corpus Striatum/physiology , Cricetinae , Cricetulus , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Space/physiology , Fear/psychology , Gene Expression Profiling , Hippocampus/physiology , Humans , Male , Maze Learning , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Prefrontal Cortex/physiology , Rats , Rats, Sprague-Dawley
12.
Handb Exp Pharmacol ; (208): 121-66, 2012.
Article in English | MEDLINE | ID: mdl-22222698

ABSTRACT

The muscarinic cholinergic system constitutes an important part of the neuronal circuitry that modulates normal cognition. Muscarinic receptor antagonists are well known to produce or exacerbate impairments in attention, learning, and memory. Conversely, both direct-acting muscarinic receptor agonists and indirect-acting muscarinic cholinergic agonists, such as acetylcholinesterase inhibitors, have shown cognition-enhancing properties, including improvements in normal cognitive function, reversal of cognitive deficits induced by muscarinic receptor antagonists, and attenuation of cognitive deficits in psychiatric and neurological disorders, such as Alzheimer's disease and schizophrenia. However, until recently, the lack of small molecule ligands that antagonize or activate specific muscarinic acetylcholine receptor (mAChR) subtypes with high selectivity has been a major obstacle in defining the relative contributions of individual mAChRs to different aspects of cognitive function and for the development of novel therapeutic agents. These limitations may be potentially overcome by the recent discovery of novel mAChR subtype-selective compounds, notably allosteric agonists and positive allosteric modulators, which exhibit greater selectivity for individual mAChR subtypes than previous mAChR orthosteric agonists. In preclinical studies, these novel ligands have shown promising efficacy in several models for the enhancement of cognition. In this chapter, we will review the muscarinic cholinergic circuitry and pharmacology of mAChR agonists and antagonists relevant to the modulation of different aspects of cognition in animals and clinical populations.


Subject(s)
Brain/drug effects , Cognition Disorders/drug therapy , Cognition/drug effects , Muscarinic Agonists/pharmacology , Nootropic Agents/pharmacology , Receptors, Muscarinic/drug effects , Signal Transduction/drug effects , Acetylcholine/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Humans , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/metabolism
13.
Neuropsychopharmacology ; 37(1): 16-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21956443

ABSTRACT

Muscarinic and nicotinic acetylcholine (ACh) receptors (mAChRs and nAChRs) are emerging as important targets for the development of novel treatments for the symptoms associated with schizophrenia. Preclinical and early proof-of-concept clinical studies have provided strong evidence that activators of specific mAChR (M(1) and M(4)) and nAChR (α(7) and α(2)ß(4)) subtypes are effective in animal models of antipsychotic-like activity and/or cognitive enhancement, and in the treatment of positive and cognitive symptoms in patients with schizophrenia. While early attempts to develop selective mAChR and nAChR agonists provided important preliminary findings, these compounds have ultimately failed in clinical development due to a lack of true subtype selectivity and subsequent dose-limiting adverse effects. In recent years, there have been major advances in the discovery of highly selective activators for the different mAChR and nAChR subtypes with suitable properties for optimization as potential candidates for clinical trials. One novel strategy has been to identify ligands that activate a specific receptor subtype through actions at sites that are distinct from the highly conserved ACh-binding site, termed allosteric sites. These allosteric activators, both allosteric agonists and positive allosteric modulators, of mAChR and nAChR subtypes demonstrate unique mechanisms of action and high selectivity in vivo, and may provide innovative treatment strategies for schizophrenia.


Subject(s)
Muscarinic Agonists/therapeutic use , Nicotinic Agonists/therapeutic use , Schizophrenia/drug therapy , Allosteric Regulation/physiology , Drug Design , Humans , Muscarinic Agonists/chemical synthesis , Nicotinic Agonists/chemical synthesis , Schizophrenia/metabolism
14.
Behav Brain Res ; 208(2): 377-82, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20006650

ABSTRACT

SPAK/STK39 is a mammalian protein kinase involved in the regulation of inorganic ion transport mechanisms known to modulate GABAergic neurotransmission in the both central and the peripheral nervous systems. We have previously shown that disruption of the gene encoding SPAK by homologous recombination in mouse embryonic stem cells results in viable mice that lack expression of the kinase. With the exception of reduced fertility, these mice do not exhibit an overt adverse phenotype. In the present study, we examine the neurological phenotype of these mice by subjecting them to an array of behavioral tests. We show that SPAK knockout mice displayed a higher nociceptive threshold than their wild-type counterparts on the hot plate and tail flick assays. SPAK knockout mice also exhibited a strong locomotor phenotype evidenced by significant deficits on the rotarod and decreased activity in open-field tests. In contrast, balance and proprioception was not affected. Finally, they demonstrated an increased anxiety-like phenotype, spending significantly longer periods of time in the dark area of the light/dark box and increased thigmotaxis in the open-field chamber. These results suggest that the kinase plays an important role in CNS function, consistent with SPAK regulating ion transport mechanisms directly involved in inhibitory neurotransmission.


Subject(s)
Behavior, Animal/physiology , Mice, Knockout/physiology , Phenotype , Protein Serine-Threonine Kinases/deficiency , Acoustic Stimulation/methods , Adaptation, Physiological/genetics , Animals , Exploratory Behavior/physiology , Functional Laterality/genetics , Hindlimb Suspension/methods , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Mice , Muscle Strength/genetics , Pain Measurement/methods , Pain Threshold/physiology , Psychomotor Performance/physiology , Reflex, Startle/genetics , Statistics, Nonparametric , Swimming/physiology
15.
Mol Pharmacol ; 76(2): 356-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19407080

ABSTRACT

Previous studies suggest that selective antagonists of specific subtypes of muscarinic acetylcholine receptors (mAChRs) may provide a novel approach for the treatment of certain central nervous system (CNS) disorders, including epileptic disorders, Parkinson's disease, and dystonia. Unfortunately, previously reported antagonists are not highly selective for specific mAChR subtypes, making it difficult to definitively establish the functional roles and therapeutic potential for individual subtypes of this receptor subfamily. The M(1) mAChR is of particular interest as a potential target for treatment of CNS disorders. We now report the discovery of a novel selective antagonist of M(1) mAChRs, termed VU0255035 [N-(3-oxo-3-(4-(pyridine-4-yl)piperazin-1-yl)propyl)-benzo[c][1,2,5]thiadiazole-4 sulfonamide]. Equilibrium radioligand binding and functional studies demonstrate a greater than 75-fold selectivity of VU0255035 for M(1) mAChRs relative to M(2)-M(5). Molecular pharmacology and mutagenesis studies indicate that VU0255035 is a competitive orthosteric antagonist of M(1) mAChRs, a surprising finding given the high level of M(1) mAChR selectivity relative to other orthosteric antagonists. Whole-cell patch-clamp recordings demonstrate that VU0255035 inhibits potentiation of N-methyl-D-aspartate receptor currents by the muscarinic agonist carbachol in hippocampal pyramidal cells. VU0255035 has excellent brain penetration in vivo and is efficacious in reducing pilocarpine-induced seizures in mice. We were surprised to find that doses of VU0255035 that reduce pilocarpine-induced seizures do not induce deficits in contextual freezing, a measure of hippocampus-dependent learning that is disrupted by nonselective mAChR antagonists. Taken together, these data suggest that selective antagonists of M(1) mAChRs do not induce the severe cognitive deficits seen with nonselective mAChR antagonists and could provide a novel approach for the treatment certain of CNS disorders.


Subject(s)
Hippocampus/metabolism , Learning/physiology , Muscarinic Antagonists/metabolism , Receptor, Muscarinic M1/metabolism , Seizures/metabolism , Animals , Binding, Competitive/drug effects , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electrophysiology , Inhibitory Concentration 50 , Male , Mice , Molecular Structure , Muscarinic Antagonists/chemistry , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Sulfonamides/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Thiadiazoles/antagonists & inhibitors , Thiadiazoles/pharmacokinetics
16.
Neurobiol Dis ; 28(1): 39-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17659877

ABSTRACT

We have previously reported CNS and locomotor deficits in KCC3 knockout mice, an animal model of agenesis of the corpus callosum associated with peripheral neuropathy (ACCPN) [Howard, H.C., Mount, D.B., Rochefort, D., Byun, N., Dupre, N., Lu, J., Fan, X., Song, L., Riviere, J.B., Prevost, C., Horst, J., Simonati, A., Lemcke, B., Welch, R., England, R., Zhan, F.Q., Mercado, A., Siesser, W.B., George, A.L., Jr., McDonald, M.P., Bouchard, J.P., Mathieu, J., Delpire, E., Rouleau, G.A., 2002. The K-Cl cotransporter KCC3 is mutant in a severe peripheral neuropathy associated with agenesis of the corpus callosum. Nat. Genet. 32, 384-392]. To assess the role of KCC3 in peripheral axon and/or myelin development and maintenance, we determined its expression and performed a detailed morphometric analysis of sciatic nerves. Sciatic nerves of juvenile wild-type mice, but not of adult, express KCC3. In the knockout, Schwann cell/myelin development appears normal at P3, but axons are swollen. At P8 and into P30, some fibers accumulate fluid periaxonally. These initial swelling pathologies are followed by axon and myelin degeneration in adult nerves, leading to reduction in nerve conduction velocity. Mutant mice also exhibit decreased sensitivity to noxious pain. This evidence for fluid-related axonopathy, which ultimately result in neurodegeneration, implicates cell volume regulation as a critical component of peripheral nerve maintenance.


Subject(s)
Axons/ultrastructure , Nerve Degeneration/pathology , Peripheral Nervous System Diseases/pathology , Sciatic Nerve/pathology , Symporters/metabolism , Animals , Blotting, Western , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Myelin Sheath/metabolism , Neural Conduction/physiology , Pain Threshold/physiology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/metabolism , Sciatic Nerve/growth & development , Sciatic Nerve/metabolism , Symporters/genetics
17.
Nat Genet ; 32(3): 384-92, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368912

ABSTRACT

Peripheral neuropathy associated with agenesis of the corpus callosum (ACCPN) is a severe sensorimotor neuropathy associated with mental retardation, dysmorphic features and complete or partial agenesis of the corpus callosum. ACCPN is transmitted in an autosomal recessive fashion and is found at a high frequency in the province of Quebec, Canada. ACCPN has been previously mapped to chromosome 15q. The gene SLC12A6 (solute carrier family 12, member 6), which encodes the K+-Cl- transporter KCC3 and maps within the ACCPN candidate region, was screened for mutations in individuals with ACCPN. Four distinct protein-truncating mutations were found: two in the French Canadian population and two in non-French Canadian families. The functional consequence of the predominant French Canadian mutation (2436delG, Thr813fsX813) was examined by heterologous expression of wildtype and mutant KCC3 in Xenopus laevis oocytes; the truncated mutant is appropriately glycosylated and expressed at the cellular membrane, where it is non-functional. Mice generated with a targeted deletion of Slc12a6 have a locomotor deficit, peripheral neuropathy and a sensorimotor gating deficit, similar to the human disease. Our findings identify mutations in SLC12A6 as the genetic lesion underlying ACCPN and suggest a critical role for SLC12A6 in the development and maintenance of the nervous system.


Subject(s)
Agenesis of Corpus Callosum , Peripheral Nervous System Diseases/genetics , Symporters/genetics , Symporters/physiology , Animals , Blotting, Southern , Brain/pathology , Canada , Chromosomes, Human, Pair 15 , Corpus Callosum/embryology , Exons , Gene Deletion , Genes, Recessive , Haplotypes , Homozygote , Humans , Immunoblotting , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Mutation , Open Reading Frames , Phenotype , Polymorphism, Genetic , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sodium-Potassium-Chloride Symporters/genetics , Spinal Cord/pathology , Time Factors , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...