Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neuron ; 112(2): 288-305.e7, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-37977151

ABSTRACT

Hunger is an internal state that not only invigorates feeding but also acts as a contextual cue for higher-order control of anticipatory feeding-related behavior. The ventral hippocampus is crucial for differentiating optimal behavior across contexts, but how internal contexts such as hunger influence hippocampal circuitry is unknown. In this study, we investigated the role of the ventral hippocampus during feeding behavior across different states of hunger in mice. We found that activity of a unique subpopulation of neurons that project to the nucleus accumbens (vS-NAc neurons) increased when animals investigated food, and this activity inhibited the transition to begin eating. Increases in the level of the peripheral hunger hormone ghrelin reduced vS-NAc activity during this anticipatory phase of feeding via ghrelin-receptor-dependent increases in postsynaptic inhibition and promoted the initiation of eating. Together, these experiments define a ghrelin-sensitive hippocampal circuit that informs the decision to eat based on internal state.


Subject(s)
Eating , Ghrelin , Mice , Animals , Ghrelin/physiology , Eating/physiology , Hippocampus , Signal Transduction/physiology , Feeding Behavior/physiology
2.
bioRxiv ; 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38014354

ABSTRACT

Dopamine release in the nucleus accumbens has been hypothesized to signal reward prediction error, the difference between observed and predicted reward, suggesting a biological implementation for reinforcement learning. Rigorous tests of this hypothesis require assumptions about how the brain maps sensory signals to reward predictions, yet this mapping is still poorly understood. In particular, the mapping is non-trivial when sensory signals provide ambiguous information about the hidden state of the environment. Previous work using classical conditioning tasks has suggested that reward predictions are generated conditional on probabilistic beliefs about the hidden state, such that dopamine implicitly reflects these beliefs. Here we test this hypothesis in the context of an instrumental task (a two-armed bandit), where the hidden state switches repeatedly. We measured choice behavior and recorded dLight signals reflecting dopamine release in the nucleus accumbens core. Model comparison based on the behavioral data favored models that used Bayesian updating of probabilistic beliefs. These same models also quantitatively matched the dopamine measurements better than non-Bayesian alternatives. We conclude that probabilistic belief computation plays a fundamental role in instrumental performance and associated mesolimbic dopamine signaling.

3.
J Neurosci ; 42(45): 8468-8476, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36351822

ABSTRACT

Prefrontal cortex (PFC) inhibitory microcircuits regulate the gain and timing of pyramidal neuron firing, coordinate neural ensemble interactions, and gate local and long-range neural communication to support adaptive cognition and contextually tuned behavior. Accordingly, perturbations of PFC inhibitory microcircuits are thought to underlie dysregulated cognition and behavior in numerous psychiatric diseases and relevant animal models. This review, based on a Mini-Symposium presented at the 2022 Society for Neuroscience Meeting, highlights recent studies providing novel insights into: (1) discrete medial PFC (mPFC) interneuron populations in the mouse brain; (2) mPFC interneuron connections with, and regulation of, long-range mPFC afferents; and (3) circuit-specific plasticity of mPFC interneurons. The contributions of such populations, pathways, and plasticity to rodent cognition are discussed in the context of stress, reward, motivational conflict, and genetic mutations relevant to psychiatric disease.


Subject(s)
Interneurons , Rodentia , Mice , Animals , Interneurons/physiology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Cognition
4.
Nat Commun ; 13(1): 339, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039510

ABSTRACT

The decision to either approach or avoid a potentially threatening environment is thought to rely upon the coordinated activity of heterogeneous neural populations in the hippocampus and prefrontal cortex (PFC). However, how this circuitry is organized to flexibly promote both approach or avoidance at different times has remained elusive. Here, we show that the hippocampal projection to PFC is composed of two parallel circuits located in the superficial or deep pyramidal layers of the CA1/subiculum border. These circuits have unique upstream and downstream connectivity, and are differentially active during approach and avoidance behaviour. The superficial population is preferentially connected to widespread PFC inhibitory interneurons, and its activation promotes exploration; while the deep circuit is connected to PFC pyramidal neurons and fast spiking interneurons, and its activation promotes avoidance. Together this provides a mechanism for regulation of behaviour during approach avoidance conflict: through two specialized, parallel circuits that allow bidirectional hippocampal control of PFC.


Subject(s)
Avoidance Learning/physiology , Behavior, Animal/physiology , Hippocampus/physiology , Prefrontal Cortex/physiology , Animals , Cholera Toxin/toxicity , Electrophysiological Phenomena , Elevated Plus Maze Test , Female , Hippocampus/anatomy & histology , Male , Mice, Inbred C57BL , Neurons/physiology , Optogenetics , Prefrontal Cortex/anatomy & histology
5.
Elife ; 102021 11 30.
Article in English | MEDLINE | ID: mdl-34845987

ABSTRACT

Projections from the basal amygdala (BA) to the ventral hippocampus (vH) are proposed to provide information about the rewarding or threatening nature of learned associations to support appropriate goal-directed and anxiety-like behaviour. Such behaviour occurs via the differential activity of multiple, parallel populations of pyramidal neurons in vH that project to distinct downstream targets, but the nature of BA input and how it connects with these populations is unclear. Using channelrhodopsin-2-assisted circuit mapping in mice, we show that BA input to vH consists of both excitatory and inhibitory projections. Excitatory input specifically targets BA- and nucleus accumbens-projecting vH neurons and avoids prefrontal cortex-projecting vH neurons, while inhibitory input preferentially targets BA-projecting neurons. Through this specific connectivity, BA inhibitory projections gate place-value associations by controlling the activity of nucleus accumbens-projecting vH neurons. Our results define a parallel excitatory and inhibitory projection from BA to vH that can support goal-directed behaviour.


Subject(s)
Amygdala/physiology , Hippocampus/physiology , Learning/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Animals , Mice , Reward
6.
Cell Rep ; 30(11): 3644-3654.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187537

ABSTRACT

The ventral subiculum (vS) of the mouse hippocampus coordinates diverse behaviors through heterogeneous populations of pyramidal neurons that project to multiple distinct downstream regions. Each of these populations of neurons is proposed to integrate a unique combination of thousands of local and long-range synaptic inputs, but the extent to which this occurs remains unknown. To address this, we employ monosynaptic rabies tracing to study the input-output relationship of vS neurons. Analysis of brain-wide inputs reveals quantitative input differences that could be explained by a combination of both the identity of the downstream target and the spatial location of the postsynaptic neurons within vS. These results support a model of combined topographical and output-defined connectivity of vS inputs. Overall, we reveal prominent heterogeneity in brain-wide inputs to the vS parallel output circuitry, providing a basis for the selective control of individual projections during behavior.


Subject(s)
Hippocampus/physiology , Neurons/physiology , Animals , Hippocampus/anatomy & histology , Imaging, Three-Dimensional , Male , Mice, Inbred C57BL , Midline Thalamic Nuclei/physiology , Rabies virus/physiology
7.
Cell Rep ; 17(2): 317-327, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705781

ABSTRACT

Correct mitochondrial distribution is critical for satisfying local energy demands and calcium buffering requirements and supporting key cellular processes. The mitochondrially targeted proteins Miro1 and Miro2 are important components of the mitochondrial transport machinery, but their specific roles in neuronal development, maintenance, and survival remain poorly understood. Using mouse knockout strategies, we demonstrate that Miro1, as opposed to Miro2, is the primary regulator of mitochondrial transport in both axons and dendrites. Miro1 deletion leads to depletion of mitochondria from distal dendrites but not axons, accompanied by a marked reduction in dendritic complexity. Disrupting postnatal mitochondrial distribution in vivo by deleting Miro1 in mature neurons causes a progressive loss of distal dendrites and compromises neuronal survival. Thus, the local availability of mitochondrial mass is critical for generating and sustaining dendritic arbors, and disruption of mitochondrial distribution in mature neurons is associated with neurodegeneration.


Subject(s)
Dendrites/genetics , Mitochondrial Proteins/genetics , Nerve Degeneration/genetics , Neurogenesis/genetics , rho GTP-Binding Proteins/genetics , Animals , Axons/metabolism , Axons/pathology , Dendrites/metabolism , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology
8.
Nat Neurosci ; 17(9): 1198-207, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108911

ABSTRACT

Repeated exposure to cocaine alters the structural and functional properties of medium spiny neurons (MSNs) in the nucleus accumbens (NAc). These changes suggest a rewiring of the NAc circuit, with an enhancement of excitatory synaptic connections onto MSNs. However, it is unknown how drug exposure alters the balance of long-range afferents onto different cell types in the NAc. Here we used whole-cell recordings, two-photon microscopy, optogenetics and pharmacogenetics to show how repeated cocaine exposure alters connectivity in the mouse NAc medial shell. Cocaine selectively enhanced amygdala innervation of MSNs expressing D1 dopamine receptors (D1-MSNs) relative to D2-MSNs. We also found that amygdala activity was required for cocaine-induced changes to behavior and connectivity. Finally, we established how heightened amygdala innervation can explain the structural and functional changes evoked by cocaine. Our findings reveal how exposure to drugs of abuse fundamentally reorganizes cell type- and input-specific connectivity in the NAc.


Subject(s)
Afferent Pathways/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Afferent Pathways/cytology , Amygdala/cytology , Amygdala/drug effects , Animals , Behavior, Animal/drug effects , Dendritic Spines/drug effects , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Mice, Mutant Strains , Neuronal Plasticity/drug effects , Optogenetics , Organ Culture Techniques , Receptors, N-Methyl-D-Aspartate/physiology , Reward
9.
Nature ; 493(7432): 411-5, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23263185

ABSTRACT

Autism spectrum disorders (ASDs) are an early onset, heterogeneous group of heritable neuropsychiatric disorders with symptoms that include deficits in social interaction skills, impaired communication abilities, and ritualistic-like repetitive behaviours. One of the hypotheses for a common molecular mechanism underlying ASDs is altered translational control resulting in exaggerated protein synthesis. Genetic variants in chromosome 4q, which contains the EIF4E locus, have been described in patients with autism. Importantly, a rare single nucleotide polymorphism has been identified in autism that is associated with increased promoter activity in the EIF4E gene. Here we show that genetically increasing the levels of eukaryotic translation initiation factor 4E (eIF4E) in mice results in exaggerated cap-dependent translation and aberrant behaviours reminiscent of autism, including repetitive and perseverative behaviours and social interaction deficits. Moreover, these autistic-like behaviours are accompanied by synaptic pathophysiology in the medial prefrontal cortex, striatum and hippocampus. The autistic-like behaviours displayed by the eIF4E-transgenic mice are corrected by intracerebroventricular infusions of the cap-dependent translation inhibitor 4EGI-1. Our findings demonstrate a causal relationship between exaggerated cap-dependent translation, synaptic dysfunction and aberrant behaviours associated with autism.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Eukaryotic Initiation Factor-4E/metabolism , Protein Biosynthesis , Synapses/metabolism , Synapses/pathology , Animals , Autistic Disorder/drug therapy , Autistic Disorder/pathology , Behavior, Animal/drug effects , Dendrites/metabolism , Dendrites/pathology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/metabolism , Female , Hippocampus/metabolism , Hydrazones , Infusions, Intraventricular , Male , Mice , Mice, Transgenic , Neostriatum/metabolism , Neuronal Plasticity , Nitro Compounds/administration & dosage , Nitro Compounds/pharmacology , Nitro Compounds/therapeutic use , Prefrontal Cortex/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , RNA Caps/metabolism , Thiazoles/administration & dosage , Thiazoles/pharmacology , Thiazoles/therapeutic use
10.
Nat Neurosci ; 15(12): 1624-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143514

ABSTRACT

We found that medium spiny neurons (MSNs) in both the direct and indirect pathways of the mouse nucleus accumbens (NAc) receive inputs from the cortex, thalamus and hippocampus. However, hippocampal inputs were much weaker onto indirect MSNs, where they contacted small spines located in the distal dendrites. This selective targeting means that these inputs must be gated by subthreshold depolarization to trigger action potentials and influence NAc output.


Subject(s)
Action Potentials/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Female , Male , Mice , Neural Pathways/cytology , Neural Pathways/physiology , Organ Culture Techniques , Subcellular Fractions/physiology
11.
Eur J Neurosci ; 32(2): 231-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20946113

ABSTRACT

Neuronal postsynaptic currents consume most of the brain's energy supply. Delineating how neurons control the distribution, morphology and function of the energy-producing mitochondria that fuel synaptic communication is therefore important for our understanding of nervous system function and pathology. Here we review recent insights into the molecular mechanisms that control activity-dependent regulation of mitochondrial trafficking, morphology and activity at excitatory synapses. We also consider some implications of this regulation for synaptic function and plasticity and discuss how this may contribute to synaptic dysfunction and signalling in neurological disease, with a focus on Alzheimer's disease.


Subject(s)
Mitochondria/metabolism , Neurons/metabolism , Synapses/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/ultrastructure , Humans , Neurons/ultrastructure , Synapses/ultrastructure , Synaptic Transmission/physiology
12.
Proc Natl Acad Sci U S A ; 107(38): 16679-84, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823221

ABSTRACT

Modification of the number of GABA(A) receptors (GABA(A)Rs) clustered at inhibitory synapses can regulate inhibitory synapse strength with important implications for information processing and nervous system plasticity and pathology. Currently, however, the mechanisms that regulate the number of GABA(A)Rs at synapses remain poorly understood. By imaging superecliptic pHluorin tagged GABA(A)R subunits we show that synaptic GABA(A)R clusters are normally stable, but that increased neuronal activity upon glutamate receptor (GluR) activation results in their rapid and reversible dispersal. This dispersal correlates with increases in the mobility of single GABA(A)Rs within the clusters as determined using single-particle tracking of GABA(A)Rs labeled with quantum dots. GluR-dependent dispersal of GABA(A)R clusters requires Ca(2+) influx via NMDA receptors (NMDARs) and activation of the phosphatase calcineurin. Moreover, the dispersal of GABA(A)R clusters and increased mobility of individual GABA(A)Rs are dependent on serine 327 within the intracellular loop of the GABA(A)R γ2 subunit. Thus, NMDAR signaling, via calcineurin and a key GABA(A)R phosphorylation site, controls the stability of synaptic GABA(A)Rs, with important implications for activity-dependent control of synaptic inhibition and neuronal plasticity.


Subject(s)
Receptors, GABA-A/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Amino Acid Substitution , Animals , Calcineurin/physiology , Calcium Signaling , Cells, Cultured , Glutamic Acid/metabolism , Multiprotein Complexes , Mutagenesis, Site-Directed , Neuronal Plasticity , Neurons/physiology , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Signal Transduction , Transfection
13.
Neuron ; 65(1): 53-65, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-20152113

ABSTRACT

The density of GABA(A) receptors (GABA(A)Rs) at synapses regulates brain excitability, and altered inhibition may contribute to Huntington's disease, which is caused by a polyglutamine repeat in the protein huntingtin. However, the machinery that delivers GABA(A)Rs to synapses is unknown. We demonstrate that GABA(A)Rs are trafficked to synapses by the kinesin family motor protein 5 (KIF5). We identify the adaptor linking the receptors to KIF5 as the huntingtin-associated protein 1 (HAP1). Disrupting the HAP1-KIF5 complex decreases synaptic GABA(A)R number and reduces the amplitude of inhibitory postsynaptic currents. When huntingtin is mutated, as in Huntington's disease, GABA(A)R transport and inhibitory synaptic currents are reduced. Thus, HAP1-KIF5-dependent GABA(A)R trafficking is a fundamental mechanism controlling the strength of synaptic inhibition in the brain. Its disruption by mutant huntingtin may explain some of the defects in brain information processing occurring in Huntington's disease and provides a molecular target for therapeutic approaches.


Subject(s)
Kinesins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Huntingtin Protein , Huntington Disease/metabolism , Kinesins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Patch-Clamp Techniques , Peptides/genetics , Peptides/metabolism , Protein Isoforms/genetics , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Synapses/ultrastructure , Synaptic Transmission/physiology
14.
Trends Cell Biol ; 20(2): 102-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20006503

ABSTRACT

Mitochondria play an essential role in ATP generation, calcium buffering and apoptotic signalling. In neurons, the transport of mitochondria to specific locations where they are needed has emerged as an important process for correct nerve cell function. Recent studies have shed light on the mechanisms that control mitochondrial transport and localization in neurons. We describe the machinery that is important for constitutive transport of mitochondria throughout the cell, and highlight recent advances in our understanding of how signalling pathways can converge on this machinery and allow for rapid activity-dependent control of mitochondrial trafficking and localization. Regulation of mitochondrial trafficking might work in concert with mitochondrial tethering systems to give precise control of mitochondrial delivery and localization to regions of high energy and calcium buffering requirements within neurons.


Subject(s)
Energy Metabolism/physiology , Mitochondria/metabolism , Nervous System/metabolism , Neurons/metabolism , Animals , Biological Transport/physiology , Calcium Signaling/physiology , Humans , Mitochondria/ultrastructure , Nervous System/ultrastructure , Neurons/ultrastructure , Protein Transport/physiology , Signal Transduction/physiology
15.
Neuron ; 61(4): 541-55, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19249275

ABSTRACT

Energy use, mainly to reverse ion movements in neurons, is a fundamental constraint on brain information processing. Trafficking of mitochondria to locations in neurons where there are large ion fluxes is essential for powering neural function. Mitochondrial trafficking is regulated by Ca2+ entry through ionotropic glutamate receptors, but the underlying mechanism is unknown. We show that the protein Miro1 links mitochondria to KIF5 motor proteins, allowing mitochondria to move along microtubules. This linkage is inhibited by micromolar levels of Ca2+ binding to Miro1. With the EF hand domains of Miro1 mutated to prevent Ca2+ binding, Miro1 could still facilitate mitochondrial motility, but mitochondrial stopping induced by glutamate or neuronal activity was blocked. Activating neuronal NMDA receptors with exogenous or synaptically released glutamate led to Miro1 positioning mitochondria at the postsynaptic side of synapses. Thus, Miro1 is a key determinant of how energy supply is matched to energy usage in neurons.


Subject(s)
Drosophila Proteins/physiology , Mitochondria/physiology , Receptors, Calcium-Sensing/physiology , Receptors, Glutamate/physiology , Synapses/physiology , rho GTP-Binding Proteins/physiology , Animals , Calcium Radioisotopes , Calcium Signaling/physiology , Cells, Cultured , Dendrites/physiology , Electrophysiology , Energy Metabolism/physiology , Glutathione Transferase/genetics , Glutathione Transferase/physiology , Immunoprecipitation , Kinesins/genetics , Kinesins/physiology , Neurons/physiology , Neurons/ultrastructure , Rats
16.
Mol Cell Neurosci ; 40(3): 301-12, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19103291

ABSTRACT

The transport of mitochondria to specific neuronal locations is critical to meet local cellular energy demands and for buffering intracellular calcium. A critical role for kinesin motor proteins in mitochondrial transport in neurons has been demonstrated. Currently however the molecular mechanisms that underlie the recruitment of motor proteins to mitochondria, and how this recruitment is regulated remain unclear. Here we show that a protein trafficking complex comprising the adaptor protein Grif-1 and the atypical GTPase Miro1 can be detected in mammalian brain where it is localised to neuronal mitochondria. Increasing Miro1 expression levels recruits Grif-1 to mitochondria. This results in an enhanced transport of mitochondria towards the distal ends of neuronal processes. Uncoupling Grif-1 recruitment to mitochondria by expressing a Grif-1/Miro1 binding fragment dramatically reduces mitochondrial transport into neuronal processes. Altering Miro1 function by mutating its first GTPase domain affects Miro's ability to recruit Grif-1 to mitochondria and in addition alters mitochondrial distribution and shape along neuronal processes. These data suggest that Miro1 and the kinesin adaptor Grif-1 play an important role in regulating mitochondrial transport in neurons.


Subject(s)
Carrier Proteins/metabolism , GTP Phosphohydrolases/metabolism , Hippocampus/cytology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Membrane Transport Proteins , Mitochondria/ultrastructure , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Molecular Motor Proteins/metabolism , Neurons/cytology , Rats , Receptors, Cell Surface , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/genetics
17.
Soc Neurosci ; 1(3-4): 259-69, 2006.
Article in English | MEDLINE | ID: mdl-18633792

ABSTRACT

Turner syndrome (TS) is a chromosomal disorder of X-monosomy in females. A minority have impaired social responsiveness, poor discrimination of facial emotions (especially fear), and abnormal amygdala-cortical connectivity. We tested the hypothesis that abnormal gaze fixation, especially with the eye region of faces, would be associated with these features, in a similar pattern to that seen in subjects with autism. Furthermore, since these features tend to be more striking in TS women whose X chromosome is maternal in origin, we also predicted that there may be a difference within the Turner's group according to parental origin of the single X. Adults with 45,X karyotype and age and IQ matched 46,XX women were recruited and tested. Facial fear recognition was significantly worse in 45,X females than controls, but there were no group differences according to parental origin of their single X chromosome. Subsequently, we tested 45,X and 46,XX women using a remote eye-tracking device, as they viewed photographs of emotional human faces. Striking differences in scanpaths were found between the TS and controls, and within the TS group, but not according to parental origin of the X chromosome. These findings provide novel evidence for abnormal face processing in some women with TS, and indicate a potential neural mechanism underlying the difficulties in some key aspects of social cognition.


Subject(s)
Eye Movements/physiology , Fear/physiology , Fear/psychology , Recognition, Psychology/physiology , Turner Syndrome/physiopathology , Turner Syndrome/psychology , Adolescent , Adult , Amygdala/physiology , Facial Expression , Female , Humans , Middle Aged , Photic Stimulation/methods
18.
Plant Physiol ; 137(1): 104-16, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15618420

ABSTRACT

The trafficking and function of cell surface proteins in eukaryotic cells may require association with detergent-resistant sphingolipid- and sterol-rich membrane domains. The aim of this work was to obtain evidence for lipid domain phenomena in plant membranes. A protocol to prepare Triton X-100 detergent-resistant membranes (DRMs) was developed using Arabidopsis (Arabidopsis thaliana) callus membranes. A comparative proteomics approach using two-dimensional difference gel electrophoresis and liquid chromatography-tandem mass spectrometry revealed that the DRMs were highly enriched in specific proteins. They included eight glycosylphosphatidylinositol-anchored proteins, several plasma membrane (PM) ATPases, multidrug resistance proteins, and proteins of the stomatin/prohibitin/hypersensitive response family, suggesting that the DRMs originated from PM domains. We also identified a plant homolog of flotillin, a major mammalian DRM protein, suggesting a conserved role for this protein in lipid domain phenomena in eukaryotic cells. Lipid analysis by gas chromatography-mass spectrometry showed that the DRMs had a 4-fold higher sterol-to-protein content than the average for Arabidopsis membranes. The DRMs were also 5-fold increased in sphingolipid-to-protein ratio. Our results indicate that the preparation of DRMs can yield a very specific set of membrane proteins and suggest that the PM contains phytosterol and sphingolipid-rich lipid domains with a specialized protein composition. Our results also suggest a conserved role of lipid modification in targeting proteins to both the intracellular and extracellular leaflet of these domains. The proteins associated with these domains provide important new experimental avenues into understanding plant cell polarity and cell surface processes.


Subject(s)
Arabidopsis/chemistry , Membrane Microdomains/chemistry , Arabidopsis/ultrastructure , Detergents , Gene Expression , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...