Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 18: 1389335, 2024.
Article in English | MEDLINE | ID: mdl-38665372

ABSTRACT

This mini review investigates the importance of GABAergic interneurons for the network function of human-induced pluripotent stem cells (hiPSC)-derived brain organoids. The presented evidence suggests that the abundance, diversity and three-dimensional cortical organization of GABAergic interneurons are the primary elements responsible for the creation of synchronous neuronal firing patterns. Without intricate inhibition, coupled oscillatory patterns cannot reach a sufficient complexity to transfer spatiotemporal information constituting physiological network function. Furthermore, human-specific brain network function seems to be mediated by a more complex and interconnected inhibitory structure that remains developmentally flexible for a longer period when compared to rodents. This suggests that several characteristics of human brain networks cannot be captured by rodent models, emphasizing the need for model systems like organoids that adequately mimic physiological human brain function in vitro.

2.
Neurosci Lett ; 816: 137509, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37802417

ABSTRACT

Latent spinal sensitization is one key mechanism developing at the early stage of chronic low back pain (LBP). TRPM3-mediated calcium transients of dorsal root ganglia (DRG) neurons are considered critical presynaptic signals involved in this latent sensitization. However, postsynaptic consequences in input laminae of the spinal cord have not been addressed so far. Here, by electrophysiological recordings in acute spinal cord slices from adult rats, we show that perfusion of the TRPM3 agonist pregnenolone sulfate (PregS) induced a significant increase in the frequency but not amplitude of spontaneous postsynaptic currents in lamina I and II neurons. This frequency increase started slowly during PregS perfusion but was reversible following washout. This result is consistent with a presynaptic action of the neurosteroid PregS, indicating the presynaptic expression of functional TRPM3 in the superficial dorsal horn of adult rats. Thus, PregS-induced TRPM3 activation enhances spinal synaptic strength, implying a mediating role of TRPM3 between neuroendocrine and nociceptive signaling, which might as well exist in chronic LBP primed by chronic stress that promotes the biosynthesis of PregS.


Subject(s)
Low Back Pain , TRPM Cation Channels , Rats , Animals , Low Back Pain/metabolism , Neurons/metabolism , Spine , Ganglia, Spinal/metabolism , TRPM Cation Channels/metabolism
3.
Cell Death Dis ; 13(10): 887, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36270985

ABSTRACT

In humans, most neurons are born during embryonic development and have to persist throughout the entire lifespan of an individual. Thus, human neurons have to develop elaborate survival strategies to protect against accidental cell death. We set out to decipher the developmental adaptations resulting in neuronal resilience. We demonstrate that, during the time course of maturation, human neurons install a complex and complementary anti-apoptotic signaling network. This includes i.) a downregulation of central proteins of the intrinsic apoptosis pathway including several caspases, ii.) a shift in the ratio of pro- and anti-apoptotic BCL-2 family proteins, and iii.) an elaborate regulatory network resulting in upregulation of the inhibitor of apoptosis protein (IAP) XIAP. Together, these adaptations strongly increase the threshold for apoptosis initiation when confronted with a wide range of cellular stressors. Our results highlight how human neurons are endowed with complex and redundant preemptive strategies to protect against stress and cell death.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Caspases/metabolism , Apoptosis/physiology , Cell Death , Inhibitor of Apoptosis Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
4.
Sci Adv ; 7(47): eabh2399, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34788104

ABSTRACT

Alcohol-dependent patients commonly show impairments in executive functions that facilitate craving and can lead to relapse. However, the molecular mechanisms leading to executive dysfunction in alcoholism are poorly understood, and new effective pharmacological treatments are desired. Here, using a bidirectional neuromodulation approach, we demonstrate a causal link between reduced prefrontal mGluR2 function and both impaired executive control and alcohol craving. A neuron-specific prefrontal mGluR2 knockdown in rats generated a phenotype of reduced cognitive flexibility and excessive alcohol seeking. Conversely, virally restoring prefrontal mGluR2 levels in alcohol-dependent rats rescued these pathological behaviors. In the search for a pharmacological intervention with high translational potential, psilocybin was capable of restoring mGluR2 expression and reducing relapse behavior. Last, we propose a FDG-PET biomarker strategy to identify mGluR2 treatment-responsive individuals. In conclusion, we identified a common molecular pathological mechanism for both executive dysfunction and alcohol craving and provided a personalized mGluR2 mechanism-based intervention strategy for medication development for alcoholism.

5.
J Neurosci ; 41(11): 2406-2419, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33531416

ABSTRACT

Extinction learning suppresses conditioned reward responses and is thus fundamental to adapt to changing environmental demands and to control excessive reward seeking. The medial prefrontal cortex (mPFC) monitors and controls conditioned reward responses. Abrupt transitions in mPFC activity anticipate changes in conditioned responses to altered contingencies. It remains, however, unknown whether such transitions are driven by the extinction of old behavioral strategies or by the acquisition of new competing ones. Using in vivo multiple single-unit recordings of mPFC in male rats, we studied the relationship between single-unit and population dynamics during extinction learning, using alcohol as a positive reinforcer in an operant conditioning paradigm. To examine the fine temporal relation between neural activity and behavior, we developed a novel behavioral model that allowed us to identify the number, onset, and duration of extinction-learning episodes in the behavior of each animal. We found that single-unit responses to conditioned stimuli changed even under stable experimental conditions and behavior. However, when behavioral responses to task contingencies had to be updated, unit-specific modulations became coordinated across the whole population, pushing the network into a new stable attractor state. Thus, extinction learning is not associated with suppressed mPFC responses to conditioned stimuli, but is anticipated by single-unit coordination into population-wide transitions of the internal state of the animal.SIGNIFICANCE STATEMENT The ability to suppress conditioned behaviors when no longer beneficial is fundamental for the survival of any organism. While pharmacological and optogenetic interventions have shown a critical involvement of the mPFC in the suppression of conditioned responses, the neural dynamics underlying such a process are still largely unknown. Combining novel analysis tools to describe behavior, single-neuron response, and population activity, we found that widespread changes in neuronal firing temporally coordinate across the whole mPFC population in anticipation of behavioral extinction. This coordination leads to a global transition in the internal state of the network, driving extinction of conditioned behavior.


Subject(s)
Behavior, Animal/physiology , Extinction, Psychological/physiology , Prefrontal Cortex/physiology , Reward , Animals , Conditioning, Operant , Learning/physiology , Male , Neurons/physiology , Rats , Rats, Wistar
7.
Commun Biol ; 4(1): 59, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420383

ABSTRACT

The NMDA receptor-mediated Ca2+ signaling during simultaneous pre- and postsynaptic activity is critically involved in synaptic plasticity and thus has a key role in the nervous system. In GRIN2-variant patients alterations of this coincidence detection provoked complex clinical phenotypes, ranging from reduced muscle strength to epileptic seizures and intellectual disability. By using our gene-targeted mouse line (Grin2aN615S), we show that voltage-independent glutamate-gated signaling of GluN2A-containing NMDA receptors is associated with NMDAR-dependent audiogenic seizures due to hyperexcitable midbrain circuits. In contrast, the NMDAR antagonist MK-801-induced c-Fos expression is reduced in the hippocampus. Likewise, the synchronization of theta- and gamma oscillatory activity is lowered during exploration, demonstrating reduced hippocampal activity. This is associated with exploratory hyperactivity and aberrantly increased and dysregulated levels of attention that can interfere with associative learning, in particular when relevant cues and reward outcomes are disconnected in space and time. Together, our findings provide (i) experimental evidence that the inherent voltage-dependent Ca2+ signaling of NMDA receptors is essential for maintaining appropriate responses to sensory stimuli and (ii) a mechanistic explanation for the neurological manifestations seen in the NMDAR-related human disorders with GRIN2 variant-meidiated intellectual disability and focal epilepsy.


Subject(s)
Calcium Signaling , Cognitive Dysfunction/genetics , Epilepsy, Reflex/genetics , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Association Learning , Attention Deficit Disorder with Hyperactivity/genetics , Hippocampus/metabolism , Mice , Proto-Oncogene Proteins c-fos/metabolism , Spatial Memory
8.
J Neurochem ; 157(3): 666-683, 2021 05.
Article in English | MEDLINE | ID: mdl-33125726

ABSTRACT

Fragile X syndrome (FXS), the most common inherited cause of intellectual disability, results from silencing of the fragile X mental retardation gene 1 (FMR1). The analyses of FXS patients' brain autopsies revealed an increased density of immature dendritic spines in cortical areas. We hypothesize that the small GTPase Arf6, an actin regulator critical for the development of glutamatergic synapses and dendritic spines, is implicated in FXS. Here, we determined the fraction of active, GTP-bound Arf6 in cortical neuron cultures and synaptoneurosomes from Fmr1 knockout mice, measured actin polymerization in neurons expressing Arf6 mutants with variant GTP- or GDP-binding properties, and recorded hippocampal long-term depression induced by metabotropic glutamate receptors (mGluR-LTD) in acute brain slices. We detected a persistently elevated Arf6 activity, a loss of Arf6 sensitivity to synaptic stimulation and an increased Arf6-dependent dendritic actin polymerization in mature Fmr1 knockout neurons. Similar imbalances in Arf6-GTP levels and actin filament assembly were caused in wild-type neurons by RNAi-mediated depletion of the postsynaptic Arf6 guanylate exchange factors IQSEC1 (BRAG2) or IQSEC2 (BRAG1). Targeted deletion of Iqsec1 in hippocampal neurons of 3-week-old mice interfered with mGluR-LTD in wild-type, but not in Fmr1 knockout mice. Collectively, these data suggest an aberrant Arf6 regulation in Fmr1 knockout neurons with consequences for the actin cytoskeleton, spine morphology, and synaptic plasticity. Moreover, FXS and syndromes caused by genetic variants in IQSEC1 and IQSEC2 share intellectual disabilities and developmental delay as main symptoms. Therefore, dysregulation of Arf6 may contribute to the cognitive impairment in FXS.


Subject(s)
ADP-Ribosylation Factors/metabolism , Fragile X Syndrome/genetics , ADP-Ribosylation Factor 6 , Actin Cytoskeleton/metabolism , Animals , Dendritic Spines/ultrastructure , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Triphosphate/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , RNA Interference , Receptors, Metabotropic Glutamate/metabolism , Synaptosomes/metabolism
9.
Sci Rep ; 7(1): 17040, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213058

ABSTRACT

Mechanisms underlying information storage have been depicted for global cell-wide and pathway-specific synaptic plasticity. Yet, little is known how these forms of plasticity interact to enhance synaptic competition and network stability. We examined synaptic interactions between apical and basal dendrites of CA1 pyramidal neurons in mouse hippocampal slices. Bursts (50 Hz) of three action potentials (AP-bursts) paired with preceding presynaptic stimulation in stratum radiatum specifically led to LTP of the paired pathway in adult mice (P75). At adolescence (P28), an increase in burst frequency (>50 Hz) was required to gain timing-dependent LTP. Surprisingly, paired radiatum and unpaired oriens pathway potentiated, unless the pre-post delay was shortened from 10 to 5 ms, which selectively potentiated paired radiatum pathway, since unpaired oriens pathway decreased back to baseline. Conversely, the exact same 5 ms pairing in stratum oriens potentiated both pathways, as did AP-bursts alone, which potentiated synaptic efficacy as well as current-evoked postsynaptic spiking. L-type voltage-gated Ca2+ channels were involved in mediating synaptic potentiation in oriens, whereas NMDA and adenosine receptors counteracted unpaired stratum oriens potentiation following pairing in stratum radiatum. This asymmetric plasticity uncovers important insights into alterations of synaptic efficacy and intrinsic neuronal excitability for pathways that convey hippocampal and extra-hippocampal information.


Subject(s)
CA1 Region, Hippocampal/metabolism , Long-Term Potentiation , Action Potentials , Animals , CA1 Region, Hippocampal/cytology , Calcium Channels, L-Type/metabolism , Electric Stimulation , Excitatory Postsynaptic Potentials , In Vitro Techniques , Mice , Receptors, GABA-B/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P1/metabolism , Synapses/physiology
10.
EMBO J ; 36(18): 2770-2789, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28790178

ABSTRACT

Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.


Subject(s)
Hippocampus/physiology , Memory, Short-Term , Protein Multimerization , Synaptic Transmission , TRPC Cation Channels/metabolism , Animals , Gene Knockout Techniques , Hippocampus/metabolism , Mass Spectrometry , Mice , Mice, Knockout , TRPC Cation Channels/genetics
11.
Front Mol Neurosci ; 10: 214, 2017.
Article in English | MEDLINE | ID: mdl-28725178

ABSTRACT

Spatial working memory (SWM) and the classical, tetanus-induced long-term potentiation (LTP) at hippocampal CA3/CA1 synapses are dependent on L-α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptors (AMPARs) containing GluA1 subunits as demonstrated by knockout mice lacking GluA1. In GluA1 knockout mice LTP and SWM deficits could be partially recovered by transgenic re-installation of full-length GluA1 in principle forebrain neurons. Here we partially restored hippocampal LTP in GluA1-deficient mice by forebrain-specific depletion of the GluA2 gene, by the activation of a hypomorphic GluA2(Q) allele and by transgenic expression of PDZ-site truncated GFP-GluA1(TG). In none of these three mouse lines, the partial LTP recovery improved the SWM performance of GluA1-deficient mice suggesting a specific function of intact GluA1/2 receptors and the GluA1 intracellular carboxyl-terminus in SWM and its associated behavior.

12.
Neurobiol Learn Mem ; 138: 281-290, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27720809

ABSTRACT

High rates of relapse after prolonged abstinence are often triggered by exposure to drug-associated cues that induce drug craving. Incubation of drug craving is a phenomenon that consists of time-dependent increases in cue-induced drug craving during withdrawal. Plasticity mechanisms in the nucleus accumbens (NAc) underlie drug-seeking responses and involve changes in excitatory synaptic transmission's efficacy. In particular, the prefrontal cortex (PFC) glutamatergic input to the NAc core has been well characterized regarding cocaine-evoked plasticity following non-contingent versus contingent exposure to cocaine or alternatively after protracted abstinence. Still, the synaptic strength during the course of withdrawal compared to drug-naïve condition is unknown, since electrophysiological characterizations are mainly performed in brain slices or focus on distinct time points during cocaine-evoked plasticity in vivo. Here we used an incubation paradigm, in which rats had extended accessed to cocaine self-administration, and underwent cue-induced reinstatement at withdrawal day 1 and 30. Longitudinal in vivo field potential recordings in awake rats showed that chronic contingent exposure to cocaine strengthened the prelimbic PFC to NAc core pathway when compared to pre-cocaine condition. This strengthening was associated with decreased paired-pulse ratios (PPR), indicative of presynaptic enhancement of glutamate release, which persisted throughout withdrawal. Moreover, both field potential increase and PPR reduction after chronic cocaine exposure correlated with the number of cocaine infusions received during training. The present results together with previous findings of withdrawal-dependent postsynaptic enhancement of the PFC-NAc core pathway, suggest an additional presynaptic strengthening that is initiated during self-administration and maintained throughout abstinence in drug-seeking rats. These cocaine-driven neuroadaptations may provide a neural substrate for maladaptive processing of cues that can ultimately trigger craving and relapse.


Subject(s)
Cocaine/administration & dosage , Craving/physiology , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Animals , Craving/drug effects , Drug-Seeking Behavior/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Self Administration
13.
Article in English | MEDLINE | ID: mdl-27818632

ABSTRACT

The glutamatergic N-methyl-D-aspartate receptor (NMDAR) is critically involved in many forms of hippocampus-dependent memory that may be enabled by synaptic plasticity. Behavioral studies with NMDAR antagonists and NMDAR subunit (GluN2) mutants revealed distinct contributions from GluN2A- and GluN2B-containing NMDARs to rapidly and slowly acquired memory performance. Furthermore, studies of synaptic plasticity, in genetically modified mice in vitro, suggest that GluN2A and GluN2B may contribute in different ways to the induction and longevity of synaptic plasticity. In contrast to the hippocampal slice preparation, in behaving mice, the afferent frequencies that induce synaptic plasticity are very restricted and specific. In fact, it is the stimulus pattern and not variations in afferent frequency that determine the longevity of long-term potentiation (LTP) in vivo. Here, we explored the contribution of GluN2A and GluN2B to LTP of differing magnitudes and persistence in freely behaving mice. We applied differing high-frequency stimulation (HFS) patterns at 100 Hz to the hippocampal CA1 region, to induce NMDAR-dependent LTP in wild-type (WT) mice, that endured for <1 h (early (E)-LTP), (LTP, 2-4 h) or >24 h (late (L)-LTP). In GluN2A-knockout (KO) mice, E-LTP (HFS, 50 pulses) was significantly reduced in magnitude and duration, whereas LTP (HFS, 2 × 50 pulses) and L-LTP (HFS, 4 × 50 pulses) were unaffected compared to responses in WT animals. By contrast, pharmacological antagonism of GluN2B in WT had no effect on E-LTP but significantly prevented LTP. E-LTP and LTP were significantly impaired by GluN2B antagonism in GluN2A-KO mice. These data indicate that the pattern of afferent stimulation is decisive for the recruitment of distinct GluN2A and GluN2B signaling pathways that in turn determine the persistency of hippocampal LTP. Whereas brief bursts of patterned stimulation preferentially recruit GluN2A and lead to weak and short-lived forms of LTP, prolonged, more intense, afferent activation recruits GluN2B and leads to robust and persistent LTP. These unique signal-response properties of GluN2A and GluN2B enable qualitative differentiation of information encoding in hippocampal synapses.

14.
Proc Natl Acad Sci U S A ; 113(11): 3024-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26903621

ABSTRACT

A major hypothesis in addiction research is that alcohol induces neuroadaptations in the mesolimbic dopamine (DA) system and that these neuroadaptations represent a key neurochemical event in compulsive drug use and relapse. Whether these neuroadaptations lead to a hypo- or hyperdopaminergic state during abstinence is a long-standing, unresolved debate among addiction researchers. The answer is of critical importance for understanding the neurobiological mechanism of addictive behavior. Here we set out to study systematically the neuroadaptive changes in the DA system during the addiction cycle in alcohol-dependent patients and rats. In postmortem brain samples from human alcoholics we found a strong down-regulation of the D1 receptor- and DA transporter (DAT)-binding sites, but D2-like receptor binding was unaffected. To gain insight into the time course of these neuroadaptations, we compared the human data with that from alcohol-dependent rats at several time points during abstinence. We found a dynamic regulation of D1 and DAT during 3 wk of abstinence. After the third week the rat data mirrored our human data. This time point was characterized by elevated extracellular DA levels, lack of synaptic response to D1 stimulation, and augmented motor activity. Further functional evidence is given by a genetic rat model for hyperdopaminergia that resembles a phenocopy of alcohol-dependent rats during protracted abstinence. In summary, we provide a new dynamic model of abstinence-related changes in the striatal DA system; in this model a hyperdopaminergic state during protracted abstinence is associated with vulnerability for relapse.


Subject(s)
Alcohol Abstinence , Alcoholism/metabolism , Dopamine/physiology , Ethanol/adverse effects , Substance Withdrawal Syndrome/metabolism , 3,4-Dihydroxyphenylacetic Acid/analysis , Adult , Aged , Animals , Benzazepines/pharmacology , Brain Chemistry , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Ethanol/toxicity , Excitatory Postsynaptic Potentials/drug effects , Female , Gene Expression Regulation , Homovanillic Acid/analysis , Humans , Male , Middle Aged , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Transgenic , Rats, Wistar , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Recurrence , Transcription, Genetic
15.
J Biol Chem ; 291(17): 9105-18, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26884337

ABSTRACT

The maturation of glutamatergic synapses in the CNS is regulated by NMDA receptors (NMDARs) that gradually change from a GluN2B- to a GluN2A-dominated subunit composition during postnatal development. Here we show that NMDARs control the activity of the small GTPase ADP-ribosylation factor 6 (Arf6) by consecutively recruiting two related brefeldin A-resistant Arf guanine nucleotide exchange factors, BRAG1 and BRAG2, in a GluN2 subunit-dependent manner. In young cortical cultures, GluN2B and BRAG1 tonically activated Arf6. In mature cultures, Arf6 was activated through GluN2A and BRAG2 upon NMDA treatment, whereas the tonic Arf6 activation was not detectable any longer. This shift in Arf6 regulation and the associated drop in Arf6 activity were reversed by a knockdown of BRAG2. Given their sequential recruitment during development, we examined whether BRAG1 and BRAG2 influence synaptic currents in hippocampal CA1 pyramidal neurons using patch clamp recordings in acute slices from mice at different ages. The number of AMPA receptor (AMPAR) miniature events was reduced by depletion of BRAG1 but not by depletion of BRAG2 during the first 2 weeks after birth. In contrast, depletion of BRAG2 during postnatal weeks 4 and 5 reduced the number of AMPAR miniature events and compromised the quantal sizes of both AMPAR and NMDAR currents evoked at Schaffer collateral synapses. We conclude that both Arf6 activation through GluN2B-BRAG1 during early development and the transition from BRAG1- to BRAG2-dependent Arf6 signaling induced by the GluN2 subunit switch are critical for the development of mature glutamatergic synapses.


Subject(s)
ADP-Ribosylation Factors/metabolism , Brefeldin A/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Synapses/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Mice , Nerve Tissue Proteins/genetics , Rats , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/physiology , Synapses/genetics
16.
Cereb Cortex ; 26(2): 647-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25270308

ABSTRACT

Activation of D4 receptors (D4Rs) has been shown to improve cognitive performance, potentially affecting synaptic strength. We investigated the D4R agonist PD 168077 (PD) in hippocampal CA1 of freely moving mice. We electrically stimulated in stratum oriens (OR) or radiatum (RAD) and evoked local field potentials (LFPs). Intraperitoneally injected PD dose-dependently and reversibly attenuated LFPs for longer time in basal (OR) than apical (RAD) dendrites. High-frequency stimulation induced LTP that was stronger and more stable in OR than RAD. LTP lasted at least 4 h during which the paired-pulse ratio remained reduced. A PD concentration not affecting synaptic transmission was sufficient to reduce LTP in OR but not in RAD. A PD concentration reducing synaptic transmission reduced the early phase LTP in OR additionally and the late phase LTP in RAD exclusively. Furthermore, cell type-specific expression of mCherry in DATCre mice generated fluorescence in dorsal CA1 that was highest in lacunosum moleculare and similar in OR/RAD, indicating that midbrain dopaminergic fibers distribute evenly in OR/RAD. Together, the D4R-mediated modulation of hippocampal synaptic transmission and plasticity is stronger in OR than RAD. This could affect information processing in CA1 neurons, since signals arriving via basal and apical afferents are distinct.


Subject(s)
Dendrites/metabolism , Hippocampus/cytology , Receptors, Dopamine D4/metabolism , Synapses/metabolism , Wakefulness/physiology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Benzamides/pharmacology , Dendrites/drug effects , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphopyruvate Hydratase/metabolism , Piperazines/pharmacology , Synapses/drug effects , Time Factors , Wakefulness/drug effects , Red Fluorescent Protein
17.
J Neurosci ; 35(27): 9946-56, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26156995

ABSTRACT

Animals are facing a complex sensory world in which only few stimuli are relevant to guide behavior. Value has to be assigned to relevant stimuli such as odors to select them over concurring information. Phasic dopamine is involved in the value assignment to stimuli in the ventral striatum. The underlying cellular mechanisms are incompletely understood. In striatal projection neurons of the ventral striatum in adult mice, we therefore examined the features and dynamics of phasic dopamine-induced synaptic plasticity and how this plasticity may modify the striatal output. Phasic dopamine is predicted to tag inputs that occur in temporal proximity. Indeed, we observed D1 receptor-dependent synaptic potentiation only when odor-like bursts and optogenetically evoked phasic dopamine release were paired within a time window of <1 s. Compatible with predictions of dynamic value assignment, the synaptic potentiation persisted after the phasic dopamine signal had ceased, but gradually reversed when odor-like bursts continued to be presented. The synaptic plasticity depended on the sensory input rate and was input specific. Importantly, synaptic plasticity amplified the firing response to a given olfactory input as the dendritic integration and the firing threshold remained unchanged during synaptic potentiation. Thus, phasic dopamine-induced synaptic plasticity can change information transfer through dynamic increases of the output of striatal projection neurons to specific sensory inputs. This plasticity may provide a neural substrate for dynamic value assignment in the striatum.


Subject(s)
Corpus Striatum/cytology , Dopamine Agents/pharmacology , Dopamine/pharmacology , Neuronal Plasticity/drug effects , Neurons/drug effects , Sensation/drug effects , Action Potentials/drug effects , Action Potentials/genetics , Afferent Pathways/drug effects , Animals , Channelrhodopsins , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lasers , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Neurons/physiology , Odorants , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Sensation/genetics , Time Factors
18.
J Neurosci ; 34(35): 11549-59, 2014 Aug 27.
Article in English | MEDLINE | ID: mdl-25164653

ABSTRACT

Phasic increases in dopamine (DA) are involved in the detection and selection of relevant sensory stimuli. The DAergic and cholinergic system dynamically interact to gate and potentiate sensory inputs to striatum. Striatal cholinergic interneurons (CINs) respond to relevant sensory stimuli with an initial burst, a firing pause, or a late burst, or a combination of these three components. CIN responses coincide with phasic firing of DAergic neurons in vivo. In particular, the late burst of CINs codes for the anticipated reward. To examine whether DAergic midbrain afferents can evoke the different CIN responses, we recorded from adult olfactory tubercle slices in the mouse ventral striatum. Olfactory inputs to striatal projection neurons were gated by the cholinergic tone. Phasic optogenetic activation of DAergic terminals evoked combinations of initial bursts, pauses, and late bursts in subsets of CINs by distinct receptor pathways. Glutamate release from midbrain afferents evoked an NMDAR-dependent initial burst followed by an afterhyperpolarization-induced pause. Phasic release of DA itself evoked acute changes in CIN firing. In particular, in CINs without an initial burst, phasic DA release evoked a pause through D2-type DA receptor activation. Independently, phasic DA activated a slow depolarizing conductance and the late burst through a D1-type DA receptor pathway. In summary, DAergic neurons elicit transient subsecond firing responses in CINs by sequential activation of NMDA, D2-type, and D1-type receptors. This fast control of striatal cholinergic tone by phasic DA provides a novel dynamic link of two transmitter systems central to the detection and selection of relevant stimuli.


Subject(s)
Cholinergic Neurons/physiology , Dopaminergic Neurons/physiology , Interneurons/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Basal Ganglia/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Patch-Clamp Techniques
19.
Addict Biol ; 18(6): 883-96, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24283978

ABSTRACT

According to the World Health Organization, about 2 billion people drink alcohol. Excessive alcohol consumption can result in alcohol addiction, which is one of the most prevalent neuropsychiatric diseases afflicting our society today. Prevention and intervention of alcohol binging in adolescents and treatment of alcoholism are major unmet challenges affecting our health-care system and society alike. Our newly formed German SysMedAlcoholism consortium is using a new systems medicine approach and intends (1) to define individual neurobehavioral risk profiles in adolescents that are predictive of alcohol use disorders later in life and (2) to identify new pharmacological targets and molecules for the treatment of alcoholism. To achieve these goals, we will use omics-information from epigenomics, genetics transcriptomics, neurodynamics, global neurochemical connectomes and neuroimaging (IMAGEN; Schumann et al. ) to feed mathematical prediction modules provided by two Bernstein Centers for Computational Neurosciences (Berlin and Heidelberg/Mannheim), the results of which will subsequently be functionally validated in independent clinical samples and appropriate animal models. This approach will lead to new early intervention strategies and identify innovative molecules for relapse prevention that will be tested in experimental human studies. This research program will ultimately help in consolidating addiction research clusters in Germany that can effectively conduct large clinical trials, implement early intervention strategies and impact political and healthcare decision makers.


Subject(s)
Alcoholism/genetics , Behavior, Addictive/genetics , Biomedical Research/methods , Genetic Predisposition to Disease/genetics , Models, Biological , Systems Biology , Adolescent , Alcohol Drinking/genetics , Alcohol Drinking/metabolism , Alcohol Drinking/therapy , Alcoholism/metabolism , Alcoholism/therapy , Animals , Behavior, Addictive/metabolism , Binge Drinking/genetics , Binge Drinking/prevention & control , Brain/drug effects , Brain/metabolism , Databases as Topic , Epigenomics , Ethanol/pharmacology , Gene Expression Profiling , Gene-Environment Interaction , Genome-Wide Association Study , Germany , Humans , Induced Pluripotent Stem Cells , Interdisciplinary Communication , Neurobiology , Neuroimaging , Polymorphism, Single Nucleotide/genetics , Precision Medicine/methods , Rats , Reward , Secondary Prevention , Transcriptome
20.
Article in English | MEDLINE | ID: mdl-23643674

ABSTRACT

NMDA receptor (NMDAR) antagonists like ketamine and MK-801 possess remarkable antidepressant effects with fast onset. However, they over-stimulate the retrosplenial cortex, evoking psychosis-like effects and neuronal injury, revealed by de novo induction of the heat shock protein 70 (Hsp70). Moreover, early in the development MK-801 triggers widespread cortical apoptosis, inducing extensive caspase-3 expression. Altogether these data raise strong concerns on the clinical applicability of NMDAR antagonist therapies. Therefore, the development of novel therapeutics targeting more specifically NMDAR to avoid psychotomimetic effects is necessary. Here we investigated a GluN2B (NR2B) antagonist in behavioral and neurotoxicity paradigms in rats to assess its potential as possible alternative to unspecific NMDA receptor antagonists. We found that treatment with the GluN2B specific antagonist Ro 25-6981 evoked robust antidepressant-like effects. Moreover, Ro 25-6981 did not cause hyperactivity as displayed after treatment with unspecific NMDAR antagonists, a correlate of psychosis-like effects in rodents. Additionally, Ro 25-6981, unlike MK-801, did not induce caspase-3 and HSP70 expression, markers of neurotoxicity in the perinatal and adult brain, respectively. Moreover, unexpectedly, in the adult retrosplenial cortex Ro 25-6981 pretreatment significantly reduced MK-801-triggered neurotoxicity. Our results suggest that GluN2B antagonists may represent valuable alternatives to unspecific NMDAR antagonists with robust antidepressant efficacy and a more favorable side-effect profile.


Subject(s)
Antidepressive Agents/pharmacology , Nerve Degeneration/chemically induced , Phenols/pharmacology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Age Factors , Animals , Caspase 3/metabolism , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Drug Interactions , Female , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , HSP70 Heat-Shock Proteins/metabolism , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Male , Mice , Motor Activity/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...