Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Neuron ; 110(15): 2409-2421.e3, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35679860

ABSTRACT

The action potential is a fundamental unit of neural computation. Even though significant advances have been made in recording large numbers of individual neurons in animal models, translation of these methodologies to humans has been limited because of clinical constraints and electrode reliability. Here, we present a reliable method for intraoperative recording of dozens of neurons in humans using the Neuropixels probe, yielding up to ∼100 simultaneously recorded single units. Most single units were active within 1 min of reaching target depth. The motion of the electrode array had a strong inverse correlation with yield, identifying a major challenge and opportunity to further increase the probe utility. Cell pairs active close in time were spatially closer in most recordings, demonstrating the power to resolve complex cortical dynamics. Altogether, this approach provides access to population single-unit activity across the depth of human neocortex at scales previously only accessible in animal models.


Subject(s)
Neocortex , Neurons , Action Potentials/physiology , Electrodes , Electrodes, Implanted , Humans , Neurons/physiology , Reproducibility of Results
2.
Cell Rep Methods ; 2(3)2022 03 28.
Article in English | MEDLINE | ID: mdl-35445205

ABSTRACT

Lesioning and neurophysiological studies have facilitated the elucidation of cortical functions and mechanisms of functional recovery following injury. Clinical translation of such studies is contingent on their employment in non-human primates (NHPs), yet tools for monitoring and modulating cortical physiology are incompatible with conventional lesioning techniques. To address these challenges, we developed a toolbox validated in seven macaques. We introduce the photothrombotic method for inducing focal cortical lesions, a quantitative model for designing experiment-specific lesion profiles and optical coherence tomography angiography (OCTA) for large-scale (~5 cm2) monitoring of vascular dynamics. We integrate these tools with our electrocorticographic array for large-scale monitoring of neural dynamics and testing stimulation-based interventions. Advantageously, this versatile toolbox can be incorporated into established chronic cranial windows. By combining optical and electrophysiological techniques in the NHP cortex, we can enhance our understanding of cortical functions, investigate functional recovery mechanisms, integrate physiological and behavioral findings, and develop neurorehabilitative treatments. MOTIVATION The primate neocortex encodes for complex functions and behaviors, the physiologies of which are yet to be fully understood. Such an understanding in both healthy and diseased states can be crucial for the development of effective neurorehabilitative strategies. However, there is a lack of a comprehensive and adaptable set of tools that enables the study of multiple physiological phenomena in healthy and injured brains. Therefore, we developed a toolbox with the capability to induce targeted cortical lesions, monitor dynamics of underlying cortical microvasculature, and record and stimulate neural activity. With this toolbox, we can enhance our understanding of cortical functions, investigate functional recovery mechanisms, test stimulation-based interventions, and integrate physiological and behavioral findings.


Subject(s)
Brain , Electric Stimulation Therapy , Animals , Brain/physiology , Primates , Macaca
3.
Sci Rep ; 10(1): 20851, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257721

ABSTRACT

Anatomic evaluation is an important aspect of many studies in neuroscience; however, it often lacks information about the three-dimensional structure of the brain. Micro-CT imaging provides an excellent, nondestructive, method for the evaluation of brain structure, but current applications to neurophysiological or lesion studies require removal of the skull as well as hazardous chemicals, dehydration, or embedding, limiting their scalability and utility. Here we present a protocol using eosin in combination with bone decalcification to enhance contrast in the tissue and then employ monochromatic and propagation phase-contrast micro-CT imaging to enable the imaging of brain structure with the preservation of the surrounding skull. Instead of relying on descriptive, time-consuming, or subjective methods, we develop simple quantitative analyses to map the locations of recording electrodes and to characterize the presence and extent of hippocampal brain lesions.


Subject(s)
Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods , X-Ray Microtomography/methods , Animals , Eosine Yellowish-(YS)/pharmacology , Hippocampus/diagnostic imaging , Imaging, Three-Dimensional/methods , Male , Prostheses and Implants , Rats , Rats, Long-Evans , Skull
4.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 3515-3518, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31946636

ABSTRACT

Ischemic stroke is a major cause of disability among adults worldwide. Despite its prevalence, few effective treatment options exist to alleviate sensory and motor dysfunctions that result from stroke. In the past, rodent models of stroke have been the primary experimental models used to develop stroke therapies. However, positive results in these studies have failed to replicate in human clinical trials, highlighting the importance of nonhuman primate (NHP) models as a preclinical step. Although there are a few NHP models of stroke, the extent of tissue damage is highly variable and dependent on surgical skill. In this study, we employed the photothrombotic stroke model in NHPs to generate controlled, reproducible ischemic lesions. Originally developed in rodents, the photothrombotic technique consists of intravenous injection of a photosensitive dye such as Rose Bengal followed by illumination of an area of interest to induce endothelial damage resulting in the formation of thrombi in the illuminated vasculature. We developed a quantitative model to predict the extent of tissue damage based on the light scattering profile of light in the cortex of NHPs. We then employed this technique in the sensorimotor cortex of two adult male Rhesus Macaques. In vivo optical coherence tomography imaging of the cortical microvasculature and subsequent histology confirmed the formation of focal cortical infarcts and demonstrated its reproducibility and ability to control the sizes and locations of light-induced ischemic lesions in the cortex of NHPs. This model has the potential to enhance our understanding of perilesional neural dynamics and can be used to develop reliable neurorehabilitative therapeutic strategies to treat stroke.


Subject(s)
Brain Ischemia , Disease Models, Animal , Stroke , Animals , Humans , Macaca mulatta , Male , Reproducibility of Results , Thrombosis , Tomography, Optical Coherence
5.
Elife ; 72018 05 29.
Article in English | MEDLINE | ID: mdl-29809133

ABSTRACT

Brain stimulation modulates the excitability of neural circuits and drives neuroplasticity. While the local effects of stimulation have been an active area of investigation, the effects on large-scale networks remain largely unexplored. We studied stimulation-induced changes in network dynamics in two macaques. A large-scale optogenetic interface enabled simultaneous stimulation of excitatory neurons and electrocorticographic recording across primary somatosensory (S1) and motor (M1) cortex (Yazdan-Shahmorad et al., 2016). We tracked two measures of network connectivity, the network response to focal stimulation and the baseline coherence between pairs of electrodes; these were strongly correlated before stimulation. Within minutes, stimulation in S1 or M1 significantly strengthened the gross functional connectivity between these areas. At a finer scale, stimulation led to heterogeneous connectivity changes across the network. These changes reflected the correlations introduced by stimulation-evoked activity, consistent with Hebbian plasticity models. This work extends Hebbian plasticity models to large-scale circuits, with significant implications for stimulation-based neurorehabilitation.


Subject(s)
Motor Cortex/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Somatosensory Cortex/physiology , Animals , Brain Waves/physiology , Connectome/methods , Dependovirus/genetics , Dependovirus/metabolism , Electrodes, Implanted , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Macaca mulatta , Male , Motor Cortex/anatomy & histology , Motor Cortex/cytology , Nerve Net/anatomy & histology , Nerve Net/cytology , Neurons/cytology , Opsins/genetics , Opsins/metabolism , Optogenetics/methods , Somatosensory Cortex/anatomy & histology , Somatosensory Cortex/cytology
6.
J Neurosci Methods ; 293: 347-358, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29042259

ABSTRACT

BACKGROUND: In non-human primate (NHP) optogenetics, infecting large cortical areas with viral vectors is often a difficult and time-consuming task. Previous work has shown that parenchymal delivery of adeno-associated virus (AAV) in the thalamus by convection-enhanced delivery (CED) can lead to large-scale transduction via axonal transport in distal areas including cortex. We used this approach to obtain widespread cortical expression of light-sensitive ion channels. NEW METHOD: AAV vectors co-expressing channelrhodopsin-2 (ChR2) and yellow fluorescent protein (YFP) genes were infused into thalamus of three rhesus macaques under MR-guided CED. After six to twelve weeks recovery, in vivo optical stimulation and single cell recording in the cortex was carried out using an optrode in anesthetized animals. Post-mortem immunostaining against YFP was used to estimate the distribution and level of expression of ChR2 in thalamus and cortex. RESULTS: Histological analysis revealed high levels of transduction in cortical layers. The patterns of expression were consistent with known thalamo-cortico-thalamic circuits. Dense expression was seen in thalamocortiocal axonal fibers in layers III, IV and VI and in pyramidal neurons in layers V and VI, presumably corticothalamic neurons. In addition we obtained reliable in vivo light-evoked responses in cortical areas with high levels of expression. COMPARISON WITH EXISTING METHODS: Thalamic CED is very efficient in achieving large expressing areas in comparison to convectional techniques both in minimizing infusion time and in minimizing damage to the brain. CONCLUSION: MR-guided CED infusion into thalamus provides a simplified approach to transduce large cortical areas by thalamo-cortico-thalamic projections in primate brain.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Macaca mulatta , Optogenetics/methods , Thalamus , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Convection , Dermoscopy , Female , Imaging, Three-Dimensional , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Magnetic Resonance Imaging , Male , Models, Animal , Neural Pathways/cytology , Neural Pathways/physiology , Photic Stimulation , Thalamus/cytology , Thalamus/diagnostic imaging , Thalamus/physiology
7.
Alcohol Clin Exp Res ; 41(2): 345-358, 2017 02.
Article in English | MEDLINE | ID: mdl-28103636

ABSTRACT

BACKGROUND: Liver damage is a serious and sometimes fatal consequence of long-term alcohol intake, which progresses from early-stage fatty liver (steatosis) to later-stage steatohepatitis with inflammation and fibrosis/necrosis. However, very little is known about earlier stages of liver disruption that may occur in problem drinkers, those who drink excessively but are not dependent on alcohol. METHODS: We examined how repeated binge-like alcohol drinking in C57BL/6 mice altered liver function, as compared with a single binge-intake session and with repeated moderate alcohol consumption. We measured a number of markers associated with early- and later-stage liver disruption, including liver steatosis, measures of liver cytochrome P4502E1 (CYP2E1) and alcohol dehydrogenase (ADH), alcohol metabolism, expression of cytokine mRNA, accumulation of 4-hydroxynonenal (4-HNE) as an indicator of oxidative stress, and alanine transaminase/aspartate transaminase as a measure of hepatocyte injury. RESULTS: Importantly, repeated binge-like alcohol drinking increased triglyceride levels in the liver and plasma, and increased lipid droplets in the liver, indicators of steatosis. In contrast, a single binge-intake session or repeated moderate alcohol consumption did not alter triglyceride levels. In addition, alcohol exposure can increase rates of alcohol metabolism through CYP2E1 and ADH, which can potentially increase oxidative stress and liver dysfunction. Intermittent, excessive alcohol intake increased liver CYP2E1 mRNA, protein, and activity, as well as ADH mRNA and activity. Furthermore, repeated, binge-like drinking, but not a single binge or moderate drinking, increased alcohol metabolism. Finally, repeated, excessive intake transiently elevated mRNA for the proinflammatory cytokine IL-1B and 4-HNE levels, but did not alter markers of later-stage liver hepatocyte injury. CONCLUSIONS: Together, we provide data suggesting that even relatively limited binge-like alcohol drinking can lead to disruptions in liver function, which might facilitate the transition to more severe forms of liver damage.


Subject(s)
Alcohol Drinking/pathology , Alcohol Drinking/psychology , Binge Drinking/pathology , Binge Drinking/psychology , Hepatitis, Alcoholic/pathology , Alanine Transaminase/blood , Alcohol Dehydrogenase/biosynthesis , Alcohol Dehydrogenase/genetics , Aldehydes/metabolism , Animals , Aspartate Aminotransferases/blood , Central Nervous System Depressants/blood , Cytochrome P-450 CYP2E1/biosynthesis , Cytochrome P-450 CYP2E1/genetics , Ethanol/blood , Interleukin-1/biosynthesis , Liver Function Tests , Male , Mice , Mice, Inbred C57BL
8.
Neuron ; 89(5): 927-39, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26875625

ABSTRACT

While optogenetics offers great potential for linking brain function and behavior in nonhuman primates, taking full advantage of that potential will require stable access for optical stimulation and concurrent monitoring of neural activity. Here we present a practical, stable interface for stimulation and recording of large-scale cortical circuits. To obtain optogenetic expression across a broad region, here spanning primary somatosensory (S1) and motor (M1) cortices, we used convection-enhanced delivery of the viral vector, with online guidance from MRI. To record neural activity across this region, we used a custom micro-electrocorticographic (µECoG) array designed to minimally attenuate optical stimuli. Lastly, we demonstrated the use of this interface to measure spatiotemporal responses to optical stimulation across M1 and S1. This interface offers a powerful tool for studying circuit dynamics and connectivity across cortical areas, for long-term studies of neuromodulation and targeted cortical plasticity, and for linking these to behavior.


Subject(s)
Brain Mapping , Cerebral Cortex/cytology , Neurons/physiology , Optogenetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Contrast Media/metabolism , Electric Stimulation , Electrodes, Implanted , Electroencephalography , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Macaca mulatta , Male , Nerve Net/physiology , Optogenetics/instrumentation , Optogenetics/methods , Photic Stimulation , Time Factors , Transduction, Genetic
9.
Front Neurosci ; 9: 487, 2015.
Article in English | MEDLINE | ID: mdl-26733798

ABSTRACT

Corticotrophin-releasing factor (CRF) is a 41 amino acid neuropeptide that coordinates adaptive responses to stress. CRF projections from neurons in the central nucleus of the amygdala (CeA) to the brainstem are of particular interest for their role in motivated behavior. To directly examine the anatomy and function of CRF neurons, we generated a BAC transgenic Crh-Cre rat in which bacterial Cre recombinase is expressed from the Crh promoter. Using Cre-dependent reporters, we found that Cre expressing neurons in these rats are immunoreactive for CRF and are clustered in the lateral CeA (CeL) and the oval nucleus of the BNST. We detected major projections from CeA CRF neurons to parabrachial nuclei and the locus coeruleus, dorsal and ventral BNST, and more minor projections to lateral portions of the substantia nigra, ventral tegmental area, and lateral hypothalamus. Optogenetic stimulation of CeA CRF neurons evoked GABA-ergic responses in 11% of non-CRF neurons in the medial CeA (CeM) and 44% of non-CRF neurons in the CeL. Chemogenetic stimulation of CeA CRF neurons induced Fos in a similar proportion of non-CRF CeM neurons but a smaller proportion of non-CRF CeL neurons. The CRF1 receptor antagonist R121919 reduced this Fos induction by two-thirds in these regions. These results indicate that CeL CRF neurons provide both local inhibitory GABA and excitatory CRF signals to other CeA neurons, and demonstrate the value of the Crh-Cre rat as a tool for studying circuit function and physiology of CRF neurons.

10.
J Neurochem ; 129(6): 1024-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24588427

ABSTRACT

The STriatal-Enriched protein tyrosine Phosphatase 61 (STEP61 ) inhibits the activity of the tyrosine kinase Fyn and dephosphorylates the GluN2B subunit of the NMDA receptor, whereas the protein kinase A phosphorylation of STEP61 inhibits the activity of the phosphatase (Pharmacol. Rev., 64, , p. 65). Previously, we found that ethanol activates Fyn in the dorsomedial striatum (DMS) leading to GluN2B phosphorylation, which, in turn, underlies the development of ethanol intake (J. Neurosci., 30, , p. 10187). Here, we tested the hypothesis that inhibition of STEP61 by ethanol is upstream of Fyn/GluN2B. We show that exposure of mice to ethanol increased STEP61 phosphorylation in the DMS, which was maintained after withdrawal and was not observed in other striatal regions. Specific knockdown of STEP61 in the DMS of mice enhanced ethanol-mediated Fyn activation and GluN2B phosphorylation, and increased ethanol intake without altering the level of water, saccharine, quinine consumption or spontaneous locomotor activity. Together, our data suggest that blockade of STEP61 activity in response to ethanol is sufficient for the activation of the Fyn/GluN2B pathway in the DMS. Being upstream of Fyn and GluN2B, inactive STEP61 in the DMS primes the induction of ethanol intake. We show that ethanol-mediated inhibition of STEP61 in the DMS leads to Fyn activation and GluN2B phosphorylation. (a) Under basal conditions, active STEP61 inhibits Fyn activity and dephosphorylates GluN2B. (b) Ethanol leads to the phosphorylation of STEP61 on a specific inhibitory site. The inhibition of STEP61 activity contributes to the activation of Fyn in response to ethanol, which, in turn, phosphorylates GluN2B. These molecular adaptations in the DMS promote ethanol drinking.


Subject(s)
Alcohol Drinking/psychology , Neostriatum/enzymology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Blotting, Western , Choice Behavior , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Immunohistochemistry , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Phosphorylation , Protein Tyrosine Phosphatases/physiology , Quinine/pharmacology , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Saccharin/pharmacology
11.
J Neurosci ; 33(36): 14369-78, 2013 Sep 04.
Article in English | MEDLINE | ID: mdl-24005290

ABSTRACT

We previously found that excessive ethanol drinking activates Fyn in the dorsomedial striatum (DMS) (Wang et al., 2010; Gibb et al., 2011). Ethanol-mediated Fyn activation in the DMS leads to the phosphorylation of the GluN2B subunit of the NMDA receptor, to the enhancement of the channel's activity, and to the development and/or maintenance of ethanol drinking behaviors (Wang et al., 2007, 2010). Protein tyrosine phosphatase α (PTPα) is essential for Fyn kinase activation (Bhandari et al., 1998), and we showed that ethanol-mediated Fyn activation is facilitated by the recruitment of PTPα to synaptic membranes, the compartment where Fyn resides (Gibb et al., 2011). Here we tested the hypothesis that PTPα in the DMS is part of the Fyn/GluN2B pathway and is thus a major contributor to the neuroadaptations underlying excessive ethanol intake behaviors. We found that RNA interference (RNAi)-mediated PTPα knockdown in the DMS reduces excessive ethanol intake and preference in rodents. Importantly, no alterations in water, saccharine/sucrose, or quinine intake were observed. Furthermore, downregulation of PTPα in the DMS of mice significantly reduces ethanol-mediated Fyn activation, GluN2B phosphorylation, and ethanol withdrawal-induced long-term facilitation of NMDAR activity without altering the intrinsic features of DMS neurons. Together, these results position PTPα upstream of Fyn within the DMS and demonstrate the important contribution of the phosphatase to the maladaptive synaptic changes that lead to excessive ethanol intake.


Subject(s)
Alcohol Drinking/metabolism , Corpus Striatum/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Adaptation, Physiological , Animals , Corpus Striatum/cytology , Corpus Striatum/metabolism , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Rats, Long-Evans , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Transcription, Genetic
12.
PLoS One ; 8(7): e68300, 2013.
Article in English | MEDLINE | ID: mdl-23869214

ABSTRACT

Human genetic association studies have shown gene variants in the α5 subunit of the neuronal nicotinic receptor (nAChR) influence both ethanol and nicotine dependence. The α5 subunit is an accessory subunit that facilitates α4* nAChRs assembly in vitro. However, it is unknown whether this occurs in the brain, as there are few research tools to adequately address this question. As the α4*-containing nAChRs are highly expressed in the ventral tegmental area (VTA) we assessed the molecular, functional and pharmacological roles of α5 in α4*-containing nAChRs in the VTA. We utilized transgenic mice α5+/+(α4YFP) and α5-/-(α4YFP) that allow the direct visualization and measurement of α4-YFP expression and the effect of the presence (α5+/+) and absence of α5 (-/-) subunit, as the antibodies for detecting the α4* subunits of the nAChR are not specific. We performed voltage clamp electrophysiological experiments to study baseline nicotinic currents in VTA dopaminergic neurons. We show that in the presence of the α5 subunit, the overall expression of α4 subunit is increased significantly by 60% in the VTA. Furthermore, the α5 subunit strengthens baseline nAChR currents, suggesting the increased expression of α4* nAChRs to be likely on the cell surface. While the presence of the α5 subunit blunts the desensitization of nAChRs following nicotine exposure, it does not alter the amount of ethanol potentiation of VTA dopaminergic neurons. Our data demonstrates a major regulatory role for the α5 subunit in both the maintenance of α4*-containing nAChRs expression and in modulating nicotinic currents in VTA dopaminergic neurons. Additionally, the α5α4* nAChR in VTA dopaminergic neurons regulates the effect of nicotine but not ethanol on currents. Together, the data suggest that the α5 subunit is critical for controlling the expression and functional role of a population of α4*-containing nAChRs in the VTA.


Subject(s)
Gene Expression Regulation , Receptors, Nicotinic/physiology , Ventral Tegmental Area/metabolism , Acetylcholine/pharmacology , Animals , Bacterial Proteins/analysis , Gene Knock-In Techniques , In Vitro Techniques , Luminescent Proteins/analysis , Male , Mice , Mice, Transgenic , Nicotine/pharmacology , Patch-Clamp Techniques , Receptors, Nicotinic/analysis , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism
13.
Nat Neurosci ; 16(8): 1111-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23792945

ABSTRACT

Relapse to alcohol abuse is an important clinical issue that is frequently caused by cue-induced drug craving. Therefore, disruption of the memory for the cue-alcohol association is expected to prevent relapse. It is increasingly accepted that memories become labile and erasable soon after their reactivation through retrieval during a memory reconsolidation process that depends on protein synthesis. Here we show that reconsolidation of alcohol-related memories triggered by the sensory properties of alcohol itself (odor and taste) activates mammalian target of rapamycin complex 1 (mTORC1) in select amygdalar and cortical regions in rats, resulting in increased levels of several synaptic proteins. Furthermore, systemic or central amygdalar inhibition of mTORC1 during reconsolidation disrupts alcohol-associated memories, leading to a long-lasting suppression of relapse. Our findings provide evidence that the mTORC1 pathway and its downstream substrates are crucial in alcohol-related memory reconsolidation and highlight this pathway as a therapeutic target to prevent relapse.


Subject(s)
Amygdala/physiopathology , Anisomycin/pharmacology , Binge Drinking/psychology , Memory/drug effects , Multiprotein Complexes/antagonists & inhibitors , Prefrontal Cortex/physiopathology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amygdala/drug effects , Animals , Anisomycin/therapeutic use , Binge Drinking/prevention & control , Conditioning, Operant/physiology , Cues , Ethanol/blood , Ethanol/chemistry , Ethanol/pharmacology , Male , Mechanistic Target of Rapamycin Complex 1 , Memory/physiology , Multiprotein Complexes/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuronal Plasticity , Odorants , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Long-Evans , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/therapeutic use , Spatial Behavior/drug effects , Spatial Behavior/physiology , TOR Serine-Threonine Kinases/physiology , Taste
14.
J Neurosci ; 32(45): 15849-58, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23136424

ABSTRACT

Uncontrolled consumption of alcohol is a hallmark of alcohol abuse disorders; however, the central molecular mechanisms underlying excessive alcohol consumption are still unclear. Here, we report that the GTP binding protein, H-Ras in the nucleus accumbens (NAc) plays a key role in neuroadaptations that underlie excessive alcohol-drinking behaviors. Specifically, acute (15 min) systemic administration of alcohol (2.5 g/kg) leads to the activation of H-Ras in the NAc of mice, which is observed even 24 h later. Similarly, rat operant self-administration of alcohol (20%) also results in the activation of H-Ras in the NAc. Using the same procedures, we provide evidence suggesting that the exchange factor GRF1 is upstream of H-Ras activation by alcohol. Importantly, we show that infection of mice NAc with lentivirus expressing a short hairpin RNA that targets the H-Ras gene produces a significant reduction of voluntary consumption of 20% alcohol. In contrast, knockdown of H-Ras in the NAc of mice did not alter water, quinine, and saccharin intake. Furthermore, using two-bottle choice and operant self-administration procedures, we show that inhibiting H-Ras activity by intra-NAc infusion of the farnesyltransferase inhibitor, FTI-276, produced a robust decrease of rats' alcohol drinking; however, sucrose consumption was unaltered. Finally, intra-NAc infusion of FTI-276 also resulted in an attenuation of seeking for alcohol. Together, these results position H-Ras as a central molecular mediator of alcohol's actions within the mesolimbic system and put forward the potential value of the enzyme as a novel target to treat alcohol use disorders.


Subject(s)
Alcohol Drinking/metabolism , Binge Drinking/metabolism , Ethanol/pharmacology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Alcohol Drinking/genetics , Animals , Binge Drinking/genetics , Choice Behavior/drug effects , Choice Behavior/physiology , Farnesyltranstransferase/antagonists & inhibitors , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Transgenic , Nucleus Accumbens/drug effects , Phosphorylation , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Rats, Long-Evans , Sucrose/pharmacology , ras-GRF1/genetics , ras-GRF1/metabolism
15.
PLoS One ; 7(4): e34559, 2012.
Article in English | MEDLINE | ID: mdl-22509321

ABSTRACT

Pavlovian fear conditioning is an associative learning paradigm in which mice learn to associate a neutral conditioned stimulus with an aversive unconditioned stimulus. In this study, we demonstrate a novel role for the transcriptional regulator Lmo4 in fear learning. LMO4 is predominantly expressed in pyramidal projection neurons of the basolateral complex of the amygdala (BLC). Mice heterozygous for a genetrap insertion in the Lmo4 locus (Lmo4gt/+), which express 50% less Lmo4 than their wild type (WT) counterparts display enhanced freezing to both the context and the cue in which they received the aversive stimulus. Small-hairpin RNA-mediated knockdown of Lmo4 in the BLC, but not the dentate gyrus region of the hippocampus recapitulated this enhanced conditioning phenotype, suggesting an adult- and brain region-specific role for Lmo4 in fear learning. Immunohistochemical analyses revealed an increase in the number of c-Fos positive puncta in the BLC of Lmo4gt/+ mice in comparison to their WT counterparts after fear conditioning. Lastly, we measured anxiety-like behavior in Lmo4gt/+ mice and in mice with BLC-specific downregulation of Lmo4 using the elevated plus maze, open field, and light/dark box tests. Global or BLC-specific knockdown of Lmo4 did not significantly affect anxiety-like behavior. These results suggest a selective role for LMO4 in the BLC in modulating learned but not unlearned fear.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amygdala/metabolism , Conditioning, Psychological/physiology , Fear/psychology , LIM Domain Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Amygdala/cytology , Amygdala/physiology , Animals , Behavior, Animal/physiology , Cues , Fear/physiology , Gene Knockdown Techniques , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Protein Transport
16.
J Clin Invest ; 122(4): 1306-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22426212

ABSTRACT

Mechanical hyperalgesia is a common and potentially disabling complication of many inflammatory and neuropathic conditions. Activation of the enzyme PKCε in primary afferent nociceptors is a major mechanism that underlies mechanical hyperalgesia, but the PKCε substrates involved downstream are not known. Here, we report that in a proteomic screen we identified the NaV1.8 sodium channel, which is selectively expressed in nociceptors, as a PKCε substrate. PKCε-mediated phosphorylation increased NaV1.8 currents, lowered the threshold voltage for activation, and produced a depolarizing shift in inactivation in wild-type - but not in PKCε-null - sensory neurons. PKCε phosphorylated NaV1.8 at S1452, and alanine substitution at this site blocked PKCε modulation of channel properties. Moreover, a specific PKCε activator peptide, ψεRACK, produced mechanical hyperalgesia in wild-type mice but not in Scn10a-/- mice, which lack NaV1.8 channels. These studies demonstrate that NaV1.8 is an important, direct substrate of PKCε that mediates PKCε-dependent mechanical hyperalgesia.


Subject(s)
Hyperalgesia/etiology , Protein Kinase C-epsilon/physiology , Protein Processing, Post-Translational , Sensory Receptor Cells/physiology , Sodium Channels/physiology , Action Potentials , Amino Acid Substitution , Animals , Cells, Cultured/drug effects , Ganglia, Spinal/cytology , Hyperalgesia/enzymology , Ion Channel Gating , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense , NAV1.8 Voltage-Gated Sodium Channel , Point Mutation , Protein Kinase C-epsilon/analysis , Protein Kinase C-epsilon/genetics , Rats , Sensory Receptor Cells/enzymology , Sodium/metabolism , Sodium Channels/analysis , Sodium Channels/chemistry , Sodium Channels/deficiency , Sodium Channels/genetics , Stress, Mechanical , Substrate Specificity
17.
J Neurosci ; 31(40): 14134-41, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976498

ABSTRACT

Previously, we showed that the mouse LIM-domain only 4 (Lmo4) gene, which encodes a protein containing two zinc-finger LIM domains that interact with various DNA-binding transcription factors, attenuates behavioral sensitivity to repeated cocaine administration. Here we show that transcription of anaplastic lymphoma kinase (Alk) is repressed by LMO4 in the striatum and that Alk promotes the development of cocaine sensitization and conditioned place preference, a measure of cocaine reward. Since LMO4 is known to interact with estrogen receptor α (ERα) at the promoters of target genes, we investigated whether Alk expression might be controlled by a similar mechanism. We found that LMO4 and ERα are associated with the Alk promoter by chromatin immunoprecipitation and that Alk is an estrogen-responsive gene in the striatum. Moreover, we show that ERα knock-out mice exhibit enhanced cocaine sensitization and conditioned place preference and an increase in Alk expression in the nucleus accumbens. These data define a novel regulatory network involved in behavioral responses to cocaine. Interestingly, sex differences in several behavioral responses to cocaine in humans and rodents have been described, and estrogen is thought to mediate some of these differences. Our data suggest that estrogen regulation of Alk may be one mechanism responsible for sexually dimorphic responses to cocaine.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Cocaine/pharmacology , Estrogen Receptor alpha/biosynthesis , Gene Targeting/methods , LIM Domain Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Reward , Transcription, Genetic/physiology , Adaptor Proteins, Signal Transducing/genetics , Anaplastic Lymphoma Kinase , Animals , Animals, Newborn , Cells, Cultured , Cocaine/metabolism , Estrogen Receptor alpha/genetics , HEK293 Cells , Humans , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription, Genetic/drug effects
18.
J Neurosci ; 30(43): 14502-12, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20980608

ABSTRACT

Spontaneous firing of ventral tegmental area (VTA) dopamine (DA) neurons provides ambient levels of DA in target areas such as the nucleus accumbens (NAc) and the prefrontal cortex (PFC). Here we report that the glial cell line-derived neurotrophic factor (GDNF), produced in one target region, the NAc, is retrogradely transported by DA neurons to the VTA where the growth factor positively regulates the spontaneous firing activity of both NAc- and PFC-projecting DA neurons in a mechanism that requires the activation of the mitogen-activated protein kinase (MAPK) pathway. We also show that the consequence of GDNF-mediated activation of the MAPK signaling cascade in the VTA is an increase in DA overflow in the NAc. Together, these results demonstrate that NAc-produced GDNF serves as a retrograde enhancer that upregulates the activity of the mesocorticolimbic DA system.


Subject(s)
Cerebral Cortex/physiology , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Limbic System/physiology , Nucleus Accumbens/metabolism , Animals , Blotting, Western , Brain Chemistry , Cloning, Molecular , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Microdialysis , Mitogen-Activated Protein Kinases/physiology , Nucleus Accumbens/physiology , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sympathectomy, Chemical , Ventral Tegmental Area/metabolism
19.
J Neurosci ; 30(26): 8830-40, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20592205

ABSTRACT

There is considerable interest in the regulation of sensorimotor gating, since deficits in this process could play a critical role in the symptoms of schizophrenia and other psychiatric disorders. Sensorimotor gating is often studied in humans and rodents using the prepulse inhibition of the acoustic startle response (PPI) model, in which an acoustic prepulse suppresses behavioral output to a startle-inducing stimulus. However, the molecular and neural mechanisms underlying PPI are poorly understood. Here, we show that a regulatory pathway involving protein phosphatase 2A (PP2A), glycogen synthase kinase 3 beta (GSK3beta), and their downstream target, the M-type potassium channel, regulates PPI. Mice (Mus musculus) carrying a hypomorphic allele of Ppp2r5delta, encoding a regulatory subunit of PP2A, show attenuated PPI. This PPP2R5delta reduction increases the phosphorylation of GSK3beta at serine 9, which inactivates GSK3beta, indicating that PPP2R5delta positively regulates GSK3beta activity in the brain. Consistently, genetic and pharmacological manipulations that reduce GSK3beta function attenuate PPI. The M-type potassium channel subunit, KCNQ2, is a putative GSK3beta substrate. Genetic reduction of Kcnq2 also reduces PPI, as does systemic inhibition of M-channels with linopirdine. Importantly, both the GSK3 inhibitor 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)1H-pyrrole-2,5-dione (SB216763) and linopirdine reduce PPI when directly infused into the medial prefrontal cortex (mPFC). Whole-cell electrophysiological recordings of mPFC neurons show that SB216763 and linopirdine have similar effects on firing, and GSK3 inhibition occludes the effects of M-channel inhibition. These data support a previously uncharacterized mechanism by which PP2A/GSK3beta signaling regulates M-type potassium channel activity in the mPFC to modulate sensorimotor gating.


Subject(s)
Auditory Perception/physiology , Brain/physiology , Glycogen Synthase Kinase 3/metabolism , Inhibition, Psychological , Potassium Channels/metabolism , Protein Phosphatase 2/metabolism , Amino Acid Sequence , Animals , Auditory Perception/drug effects , Brain/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Neurons/drug effects , Neurons/physiology , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Protein Phosphatase 2/genetics , Reflex, Startle/drug effects , Reflex, Startle/physiology , Signal Transduction
20.
Neuropsychopharmacology ; 35(6): 1363-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20164830

ABSTRACT

Cannabinoid agonists have shown some promise clinically as analgesics, in particular for cancer pain, in which they have the additional benefit of decreasing nausea. However, as for most other drugs, the long-term use of cannabinoids is limited by the development of tolerance. Several molecular mechanisms have been proposed to explain drug tolerance, including receptor downregulation. The cannabinoid 1 (CB1) receptors can be downregulated in vitro through an interaction with the G-protein-coupled receptor-associated sorting protein1, GASP1, that targets CB1 receptors for degradation after their agonist-mediated endocytosis. To investigate whether GASP1-mediated postendocytic sorting of the CB1 receptor contributes to tolerance to cannabinoid drugs in vivo, we generated a mouse with a disruption of GASP1. In wild-type mice, repeated administration of the cannabinoid agonist WIN55,212-2 promoted downregulation of CB1 receptor levels and concomitant tolerance to the effects of drug on antinociception, motor incoordination, and locomotor hypoactivity. In contrast, GASP1 knockout mice did not develop tolerance to any of these effects and showed no significant receptor downregulation. Taken together, this study provides evidence that GASP1 regulates CB1 receptor downregulation in vivo, and that postendocytic receptor trafficking has a key role in the development of tolerance to WIN55,212-2.


Subject(s)
Benzoxazines/pharmacology , Cannabinoids/pharmacology , Carrier Proteins/genetics , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Analgesics/pharmacology , Animals , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Tolerance/physiology , Endocytosis/drug effects , Endocytosis/physiology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Pain/drug therapy , Pain/genetics , Pain/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Receptor, Cannabinoid, CB1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...