Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 32(7): 1350-1364, 2022 03 30.
Article in English | MEDLINE | ID: mdl-34470044

ABSTRACT

Neurotrophins are secreted proteins that control survival, differentiation, and synaptic plasticity. While mature neurotrophins regulate these functions via tyrosine kinase signaling (Trk), uncleaved pro-neurotrophins bind preferentially to the p75 neurotrophin receptor (p75NTR) and often exert opposite effects to those of mature neurotrophins. In the amygdala, brain-derived neurotrophic factor (BDNF) enables long-term potentiation as well as fear and fear extinction learning. In the present study, we focused on the impact of mature BDNF and proBDNF signaling on long-term depression (LTD) in the lateral amygdala (LA). Hence, we conducted extracellular field potential recordings in an in vitro slice preparation and recorded LTD in cortical and thalamic afferents to the LA. LTD was unchanged by acute block of BDNF/TrkB signaling. In contrast, LTD was inhibited by blocking p75NTR signaling, by disinhibition of the proteolytic cleavage of proBDNF into mature BDNF, and by preincubation with a function-blocking anti-proBDNF antibody. Since LTD-like processes in the amygdala are supposed to be related to fear extinction learning, we locally inhibited p75NTR signaling in the amygdala during or after fear extinction training, resulting in impaired fear extinction memory. Overall, these results suggest that in the amygdala proBDNF/p75NTR signaling plays a pivotal role in LTD and fear extinction learning.


Subject(s)
Extinction, Psychological , Fear , Amygdala/metabolism , Animals , Extinction, Psychological/physiology , Fear/physiology , Learning/physiology , Mice , Neuronal Plasticity
2.
Int J Mol Sci ; 20(17)2019 Sep 03.
Article in English | MEDLINE | ID: mdl-31484392

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has previously been shown to play an important role in glutamatergic synaptic plasticity in the amygdala, correlating with cued fear learning. While glutamatergic neurotransmission is facilitated by BDNF signaling in the amygdala, its mechanism of action at inhibitory synapses in this nucleus is far less understood. We therefore analyzed the impact of chronic BDNF depletion on GABAA-mediated synaptic transmission in BDNF heterozygous knockout mice (BDNF+/-). Analysis of miniature and evoked inhibitory postsynaptic currents (IPSCs) in the lateral amygdala (LA) revealed neither pre- nor postsynaptic differences in BDNF+/- mice compared to wild-type littermates. In addition, long-term potentiation (LTP) of IPSCs was similar in both genotypes. In contrast, facilitation of spontaneous IPSCs (sIPSCs) by norepinephrine (NE) was significantly reduced in BDNF+/- mice. These results argue against a generally impaired efficacy and plasticity at GABAergic synapses due to a chronic BDNF deficit. Importantly, the increase in GABAergic tone mediated by NE is reduced in BDNF+/- mice. As release of NE is elevated during aversive behavioral states in the amygdala, effects of a chronic BDNF deficit on GABAergic inhibition may become evident in response to states of high arousal, leading to amygdala hyper-excitability and impaired amygdala function.


Subject(s)
Amygdala/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Long-Term Potentiation/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Long-Term Potentiation/genetics , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Synaptic Transmission/genetics , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
3.
Mol Neurobiol ; 56(10): 6833-6855, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30929164

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a secreted messenger molecule that is crucial for neuronal function and induction of synaptic plasticity. Although altered availability of BDNF underlies many neurological deficits and neurodegenerative disorders, secretion dynamics of endogenous BDNF are unexplored. We generated a BDNF-GFP knock-in (KiBE) mouse, in which GFP-labeled BDNF is expressed under the control of the unaltered endogenous mouse BDNF gene regulatory elements. This KiBE mouse model enables for the first time live cell imaging analysis of endogenous BDNF dynamics. We show that BDNF-GFP release and biological activity in vivo are unaffected by the GFP tag, since homozygous KiBE mice, which lack wild-type BDNF, are healthy and have a normal life expectancy. STED superresolution microscopy shows that 70% of BDNF-GFP vesicles in KiBE mouse neurites are localized in dendrites, being typically 200 nm away from synaptic release sites. Live cell imaging in hippocampal slices also reveals prominent targeting of endogenous BDNF-GFP vesicles to dendrites. Fusion pore opening and cargo release of dendritic BDNF vesicles start within 30 s after a strong depolarizing stimulus and continue for > 100 s thereafter, revealing an astonishingly delayed and prolonged release of endogenous BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dendrites/metabolism , Exocytosis , Gene Knock-In Techniques , Green Fluorescent Proteins/metabolism , Synaptic Vesicles/metabolism , Animals , Axons/metabolism , Cells, Cultured , Chromosomes, Mammalian/genetics , Gene Targeting , Genome , Hippocampus/metabolism , Mice
4.
J Biomed Mater Res B Appl Biomater ; 107(6): 1806-1813, 2019 08.
Article in English | MEDLINE | ID: mdl-30508321

ABSTRACT

The reasons for the high number of loosened metal-on-metal (MoM) hip implants are still not fully understood. Hypoxia-inducible factor 1 (HIF-1) mediated signaling pathways, which normally modulate tissue metabolism under hypoxic circumstances, could be triggered by metallic wear debris and influence bone metabolism favoring osteolysis. This may lead to early loosening of the orthopedic implants. Immunhistochemical staining of periprosthetic tissues of failed artificial hip implants showed that the concentration of HIF-1α in the surrounding tissues of failed MoM hip implants was significantly higher in comparison to failed metal-on-polyethylene (MoP) hip implants and osteoarthritic tissues. Therefore, we examined the Co2+ -uptake mechanisms and the influence of Co2+ uptake on HIF-1α stabilization. Based on cobalt mediated quenching effects, calcium imaging experiments using fura-2 showed a concentration-dependent cobalt influx in MG-63 cells, which could be inhibited by the unspecific TRPM7 channel inhibitor 2-APB (20 µM) and TRPM7 specific siRNA. Western blots confirmed a dose dependent increase of HIF-1α upon stimulation with Co2+ . This effect could be abrogated by inhibition of cobalt influx using 2-APB. This study shows that chemical hypoxia originating from HIF-1α upregulation within the periprosthetic tissue is related to cobalt wear debris and highlights TRPM7 as an important key mediator in this context. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1806-1813, 2019.


Subject(s)
Cobalt/pharmacokinetics , Gene Expression Regulation , Hip Prosthesis , Metal-on-Metal Joint Prostheses , Osteoblasts/metabolism , Protein Serine-Threonine Kinases/biosynthesis , TRPM Cation Channels/biosynthesis , Cell Hypoxia , Cell Line , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
5.
Nat Neurosci ; 21(7): 952-962, 2018 07.
Article in English | MEDLINE | ID: mdl-29950668

ABSTRACT

Functional neuroanatomy of Pavlovian fear has identified neuronal circuits and synapses associating conditioned stimuli with aversive events. Hebbian plasticity within these networks requires additional reinforcement to store particularly salient experiences into long-term memory. Here we have identified a circuit that reciprocally connects the ventral periaqueductal gray and dorsal raphe region with the central amygdala and that gates fear learning. We found that ventral periaqueductal gray and dorsal raphe dopaminergic (vPdRD) neurons encode a positive prediction error in response to unpredicted shocks and may reshape intra-amygdala connectivity via a dopamine-dependent form of long-term potentiation. Negative feedback from the central amygdala to vPdRD neurons might limit reinforcement to events that have not been predicted. These findings add a new module to the midbrain dopaminergic circuit architecture underlying associative reinforcement learning and identify vPdRD neurons as a critical component of Pavlovian fear conditioning. We propose that dysregulation of vPdRD neuronal activity may contribute to fear-related psychiatric disorders.


Subject(s)
Association Learning/physiology , Dopaminergic Neurons/physiology , Fear/physiology , Tegmentum Mesencephali/physiology , Animals , Behavior, Animal/physiology , Conditioning, Classical/physiology , Long-Term Potentiation/physiology , Male , Mice , Neural Pathways/physiology , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Tegmentum Mesencephali/cytology
6.
PLoS One ; 13(3): e0192652, 2018.
Article in English | MEDLINE | ID: mdl-29513687

ABSTRACT

The possibility to generate cardiomyocytes from pluripotent stem cells in vitro has enormous significance for basic research, disease modeling, drug development and heart repair. The concept of heart muscle reconstruction has been studied and optimized in the rat model using rat primary cardiovascular cells or xenogeneic pluripotent stem cell derived-cardiomyocytes for years. However, the lack of rat pluripotent stem cells (rPSCs) and their cardiovascular derivatives prevented the establishment of an authentic clinically relevant syngeneic or allogeneic rat heart regeneration model. In this study, we comparatively explored the potential of recently available rat embryonic stem cells (rESCs) and induced pluripotent stem cells (riPSCs) as a source for cardiomyocytes (CMs). We developed feeder cell-free culture conditions facilitating the expansion of undifferentiated rPSCs and initiated cardiac differentiation by embryoid body (EB)-formation in agarose microwell arrays, which substituted the robust but labor-intensive hanging drop (HD) method. Ascorbic acid was identified as an efficient enhancer of cardiac differentiation in both rPSC types by significantly increasing the number of beating EBs (3.6 ± 1.6-fold for rESCs and 17.6 ± 3.2-fold for riPSCs). These optimizations resulted in a differentiation efficiency of up to 20% cTnTpos rPSC-derived CMs. CMs showed spontaneous contractions, expressed cardiac markers and had typical morphological features. Electrophysiology of riPSC-CMs revealed different cardiac subtypes and physiological responses to cardio-active drugs. In conclusion, we describe rPSCs as a robust source of CMs, which is a prerequisite for detailed preclinical studies of myocardial reconstruction in a physiologically and immunologically relevant small animal model.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryoid Bodies/cytology , Feeder Cells , Myocardial Contraction , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Rats, Inbred F344 , Reproducibility of Results
7.
Front Cell Neurosci ; 8: 323, 2014.
Article in English | MEDLINE | ID: mdl-25426021

ABSTRACT

BDNF and nitric oxide signaling both contribute to plasticity at glutamatergic synapses. However, the role of combined signaling of both pathways at the same synapse is largely unknown. Using NO imaging with diaminofluoresceine in cultured hippocampal neurons we analyzed the time course of neurotrophin-induced NO signals. Application of exogenous BDNF, NT-4, and NT-3 (but not NGF) induced NO signals in the soma and in proximal dendrites of hippocampal neurons that were sensitive to NO synthase activity, TrkB signaling, and intracellular calcium elevation. The effect of NO signaling on neurotrophin secretion was analyzed in BDNF-GFP, and NT-3-GFP transfected hippocampal neurons. Exogenous application of the NO donor sodium-nitroprusside markedly inhibited neurotrophin secretion. However, endogenously generated NO in response to depolarization and neurotrophin stimulation, both did not result in a negative feedback on neurotrophin secretion. These results suggest that a negative feedback of NO signaling on synaptic secretion of neurotrophins operates only at high intracellular levels of nitric oxide that are under physiological conditions not reached by depolarization or BDNF signaling.

8.
Neurosignals ; 20(1): 35-47, 2012.
Article in English | MEDLINE | ID: mdl-22094222

ABSTRACT

The mechanisms underlying spontaneous burst activity (SBA), appearing in networks of embryonic cortical neurons at the end of the first week in vitro, remain elusive. Here we investigated the contribution of the hyperpolarization-activated cation current (I(h)) to SBA in cortical cultures of GAD67-GFP mice. I(h) current could be detected in GFP-positive large GABAergic interneurons (L-INs) and glutamatergic principal neurons (PNs) as early as DIV 5. Under current-clamp conditions, blockers of I(h) current, ZD7288 and Cs⁺, abolished the voltage sag and rebound depolarization. ZD7288 induced a hyperpolarization concomitant with an increase in the membrane input resistance in L-INs and PNs. Voltage-clamp recordings revealed I(h) as slowly activating inward current with a reversal potential close to -50 mV and a mid-activation point around -90 mV. Both, ZD7288 (1-10 µM) and Cs⁺ (1-2 mM) reduced SBA, spontaneous activity-driven Ca²âº transients, and frequency as well as amplitude of miniature GABAergic postsynaptic currents. Immunocytochemistry and Western blot demonstrated that HCN1 and HCN2 were the prevalent isoforms of HCN channels expressed in L-INs and PNs. These results suggest an important contribution of HCN channels to the maintenance of SBA in embryonic cortical cultures.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/physiology , Ion Channels/physiology , Neocortex/embryology , Neocortex/growth & development , Nerve Net/embryology , Nerve Net/growth & development , Potassium Channels/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Cells, Cultured , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neocortex/cytology , Nerve Net/cytology , Neural Conduction/physiology , Protein Isoforms/physiology
9.
PLoS One ; 6(12): e27474, 2011.
Article in English | MEDLINE | ID: mdl-22164209

ABSTRACT

Neuronal high-voltage-activated (HVA) Ca(2+) channels are rapidly inactivated by a mechanism that is termed Ca(2+)-dependent inactivation (CDI). In this study we have shown that ß-adrenergic receptor (ßAR) stimulation inhibits CDI in rat thalamocortical (TC) relay neurons. This effect can be blocked by inhibition of cAMP-dependent protein kinase (PKA) with a cell-permeable inhibitor (myristoylated protein kinase inhibitor-(14-22)-amide) or A-kinase anchor protein (AKAP) St-Ht31 inhibitory peptide, suggesting a critical role of these molecules downstream of the receptor. Moreover, inhibition of protein phosphatases (PP) with okadaic acid revealed the involvement of phosphorylation events in modulation of CDI after ßAR stimulation. Double fluorescence immunocytochemistry and pull down experiments further support the idea that modulation of CDI in TC neurons via ßAR stimulation requires a protein complex consisting of Ca(V)1.2, PKA and proteins from the AKAP family. All together our data suggest that AKAPs mediate targeting of PKA to L-type Ca(2+) channels allowing their phosphorylation and thereby modulation of CDI.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium/chemistry , Receptors, Adrenergic, beta/metabolism , Animals , COS Cells , Cerebral Cortex/pathology , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , Immunohistochemistry/methods , Neurons/metabolism , Okadaic Acid/pharmacology , Phosphorylation , Rats , Rats, Long-Evans , Signal Transduction , Thalamus/pathology , Tissue Distribution
10.
PLoS One ; 6(3): e18020, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21437203

ABSTRACT

The neuropeptide S (NPS) receptor system modulates neuronal circuit activity in the amygdala in conjunction with fear, anxiety and the expression and extinction of previously acquired fear memories. Using in vitro brain slice preparations of transgenic GAD67-GFP (Δneo) mice, we investigated the effects of NPS on neural activity in the lateral amygdala as a key region for the formation and extinction of fear memories. We are able to demonstrate that NPS augments excitatory glutamatergic synaptic input onto both projection neurons and interneurons of the lateral amygdala, resulting in enhanced spike activity of both types of cells. These effects were at least in part mediated by presynaptic mechanisms. In turn, inhibition of projection neurons by local interneurons was augmented by NPS, and subthreshold oscillations were strengthened, leading to their shift into the theta frequency range. These data suggest that the multifaceted effects of NPS on amygdaloid circuitry may shape behavior-related network activity patterns in the amygdala and reflect the peptide's potent activity in various forms of affective behavior and emotional memory.


Subject(s)
Action Potentials/drug effects , Amygdala/drug effects , Amygdala/physiology , Neuropeptides/pharmacology , Synaptic Transmission/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , In Vitro Techniques , Interneurons/drug effects , Interneurons/physiology , Mice , Patch-Clamp Techniques
11.
Eur J Neurosci ; 31(3): 439-49, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20105233

ABSTRACT

Neuronal Ca(2+) channels are rapidly inactivated by a mechanism that is termed Ca(2+)-dependent inactivation (CDI). In this study we investigated the influence of intracellular Ca(2+) release on CDI of high-voltage-activated Ca(2+) channels in rat thalamocortical relay neurons by combining voltage-clamp, Ca(2+) imaging and immunological techniques. Double-pulse protocols revealed CDI, which depended on the length of the conditioning pulses. Caffeine caused a concentration-dependent increase in CDI that was accompanied by an increase in the duration of Ca(2+) transients. Inhibition of ryanodine receptors and endoplasmic Ca(2+) pumps (by thapsigargin or cyclopiazonic acid) resulted in a reduction of CDI. In contrast, inhibition of inositol 1,4,5-tris-phosphate receptors by intracellular application of 2-aminoethoxy diphenyl borate or heparin did not influence CDI. The block of transient receptor potential channels by extracellular application of 2-aminoethoxy diphenyl borate, however, resulted in a significant reduction of CDI. The central role of L-type Ca(2+) channels was emphasized by the near-complete block of CDI by nifedipine, an effect only surpassed when Ca(2+) was replaced by Ba(2+) and chelated by 1,2-bis(o-aminophenoxy)ethane-N,N,N',N',-tetraacetic acid (BAPTA). Trains of action potential-like stimuli induced a strong reduction in high-voltage-activated Ca(2+) current amplitude, which was significantly reduced when intracellular Ca(2+) stores were made inoperative by thapsigargin or Ba(2+)/BAPTA. Western blotting revealed expression of L-type Ca(2+) channels in thalamic and hippocampal tissue but not liver tissue. In summary, these results suggest a cross-signalling between L-type Ca(2+) channels and ryanodine receptors that controls the amount of Ca(2+) influx during neuronal activity.


Subject(s)
Afferent Pathways/metabolism , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Cerebral Cortex/cytology , Neurons/physiology , Thalamus/cytology , Animals , Boron Compounds/metabolism , Calcium Channel Blockers/metabolism , Chelating Agents/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Enzyme Inhibitors/metabolism , Ion Channel Gating/physiology , Neurons/cytology , Nifedipine/metabolism , Patch-Clamp Techniques , Rats , Rats, Long-Evans , Ryanodine/metabolism , Thapsigargin/metabolism
12.
Front Cell Neurosci ; 4: 132, 2010.
Article in English | MEDLINE | ID: mdl-21267426

ABSTRACT

In thalamocortical relay (TC) neurons, G-protein-coupled receptors play an important part in the control of activity modes. A conditional Gα(q) knockout on the background of a constitutive Gα(11) knockout (Gα(q)/Gα(11) (-/-)) was used to determine the contribution of Gq/G11 family G-proteins to metabotropic serotonin (5-HT) and glutamate (Glu) function in the dorsal part of the lateral geniculate nucleus (dLGN). In control mice, current clamp recordings showed that α-m-5-HT induced a depolarization of V(rest) which was sufficient to suppress burst firing. This depolarization was concentration-dependent (100 µM: +6 ± 1 mV, n = 10; 200 µM: +10 ± 1 mV, n = 7) and had a conditioning effect on the activation of other Gα(q)-mediated pathways. The depolarization was significantly reduced in Gα(q)/Gα(11) (-/-) (100 µM: 3 ± 1 mV, n = 11; 200 µM: 5 ± 1 mV, n = 6) and was apparently insufficient to suppress burst firing. Activating Gα(q)-coupled muscarinic receptors affected the magnitude of α-m-5-HT-induced effects in a reciprocal manner. Furthermore, the depolarizing effect of mGluR1 agonists was significantly reduced in Gα(q)/Gα(11) (-/-) mice. Immunohistochemical stainings revealed binding of 5-HT(2C)R- and mGluR1α-, but not of 5-HT(2A)R-specific antibodies in the dLGN of Gα(q)/Gα(11) (-/-) mice. In conclusion, these findings demonstrate that transmitters of ascending brainstem fibers and corticofugal fibers both signal via a central element in the form of Gq/G11-mediated pathways to control activity modes in the TC system.

13.
J Neurosci ; 29(49): 15397-409, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20007464

ABSTRACT

Cytotoxic CD8(+) T cells are considered important effector cells contributing to neuronal damage in inflammatory and degenerative CNS disorders. Using time-lapse video microscopy and two-photon imaging in combination with whole-cell patch-clamp recordings, we here show that major histocompatibility class I (MHC I)-restricted neuronal antigen presentation and T cell receptor specificity determine CD8(+) T-cell locomotion and neuronal damage in culture and hippocampal brain slices. Two separate functional consequences result from a direct cell-cell contact between antigen-presenting neurons and antigen-specific CD8(+) T cells. (1) An immediate impairment of electrical signaling in single neurons and neuronal networks occurs as a result of massive shunting of the membrane capacitance after insertion of channel-forming perforin (and probably activation of other transmembrane conductances), which is paralleled by an increase of intracellular Ca(2+) levels (within <10 min). (2) Antigen-dependent neuronal apoptosis may occur independently of perforin and members of the granzyme B cluster (within approximately 1 h), suggesting that extracellular effects can substitute for intracellular delivery of granzymes by perforin. Thus, electrical silencing is an immediate consequence of MHC I-restricted interaction of CD8(+) T cells with neurons. This mechanism is clearly perforin-dependent and precedes, but is not causally linked, to neuronal cell death.


Subject(s)
CD8 Antigens/metabolism , Cell Communication/physiology , Neurons/physiology , Perforin/metabolism , T-Lymphocytes/physiology , Animals , Calcium/metabolism , Cell Death/physiology , Cell Movement/physiology , Cells, Cultured , Electric Capacitance , Genes, MHC Class I/physiology , Granzymes/genetics , Granzymes/metabolism , Hippocampus/physiology , In Vitro Techniques , Intracellular Space/physiology , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Perforin/genetics , Time Factors
14.
Proc Natl Acad Sci U S A ; 106(22): 9093-8, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19458041

ABSTRACT

Phosphatidylinositol 4-OH kinase IIIbeta (PI-4Kbeta) is involved in the regulated local synthesis of phospholipids that are crucial for trans-Golgi network (TGN)-to-plasma membrane trafficking. In this study, we show that the calcium sensor proteins calneuron-1 and calneuron-2 physically associate with PI-4Kbeta, inhibit the enzyme profoundly at resting and low calcium levels, and negatively interfere with Golgi-to-plasma membrane trafficking. At high calcium levels this inhibition is released and PI-4Kbeta is activated via a preferential association with neuronal calcium sensor-1 (NCS-1). In accord to its supposed function as a filter for subthreshold Golgi calcium transients, neuronal overexpression of calneuron-1 enlarges the size of the TGN caused by a build-up of vesicle proteins and reduces the number of axonal Piccolo-Bassoon transport vesicles, large dense core vesicles that carry a set of essential proteins for the formation of the presynaptic active zone during development. A corresponding protein knockdown has the opposite effect. The opposing roles of calneurons and NCS-1 provide a molecular switch to decode local calcium transients at the Golgi and impose a calcium threshold for PI-4Kbeta activity and vesicle trafficking.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Calcium Signaling , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , trans-Golgi Network/metabolism , 1-Phosphatidylinositol 4-Kinase/metabolism , Animals , COS Cells , Calcium/metabolism , Calcium-Binding Proteins/genetics , Chlorocebus aethiops , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Protein Transport , Rats
15.
Exp Cell Res ; 315(1): 50-66, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18992240

ABSTRACT

The bone marrow represents an easy accessible source of adult stem cells suitable for various cell based therapies. Several studies in recent years suggested the existence of pluripotent stem cells within bone marrow stem cells (BMSC) expressing marker proteins of both embryonic and tissue committed stem cells. These subpopulations were referred to as MAPC, MIAMI and VSEL-cells. Here we describe SD-BMSC (serumdeprivation-induced BMSC) which are induced as a distinct subpopulation after complete serumdeprivation. SD-BMSC are generated from small-sized nestin-positive BMSC (S-BMSC) organized as round-shaped cells in the top layer of BMSC-cultures. The generation of SD-BMSC is caused by a selective proliferation of S-BMSC and accompanied by changes in both morphology and gene expression. SD-BMSC up-regulate not only markers typical for neural stem cells like nestin and GFAP, but also proteins characteristic for embryonic cells like Oct4 and SOX2. We hypothesize, that SD-BMSC like MAPC, MIAMI and VSEL-cells represent derivatives from a single pluripotent stem cell fraction within BMSC exhibiting characteristics of embryonic and tissue committed stem cells. The complete removal of serum might offer a simple way to specifically enrich this fraction of pluripotent embryonic like stem cells in BMSC cultures.


Subject(s)
Bone Marrow Cells/cytology , Embryonic Stem Cells/cytology , Neurons/cytology , Serum/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Polarity , Cell Proliferation , Cell Shape , Cells, Cultured , Cellular Structures/metabolism , Electrophysiological Phenomena , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
16.
PLoS One ; 3(7): e2695, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18628994

ABSTRACT

Neuropeptide S (NPS) and its receptor are thought to define a set of specific brain circuits involved in fear and anxiety. Here we provide evidence for a novel, NPS-responsive circuit that shapes neural activity in the mouse basolateral amygdala (BLA) via the endopiriform nucleus (EPN). Using slice preparations, we demonstrate that NPS directly activates an inward current in 20% of EPN neurons and evokes an increase of glutamatergic excitation in this nucleus. Excitation of the EPN is responsible for a modulation of BLA activity through NPS, characterized by a general increase of GABAergic inhibition and enhancement of spike activity in a subset of BLA projection neurons. Finally, local injection of NPS to the EPN interferes with the expression of contextual, but not auditory cued fear memory. Together, these data suggest the existence of a specific NPS-responsive circuitry between EPN and BLA, likely involved in contextual aspects of fear memory.


Subject(s)
Amygdala/metabolism , Neuropeptides/chemistry , Animals , Brain Mapping , Electrophysiology , Glutamine/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Neurological , Nerve Net , Neuronal Plasticity , Neurons/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
17.
Pflugers Arch ; 456(6): 1049-60, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18350314

ABSTRACT

A genetic knock out was used to determine the specific contribution of G(q)/G(11)-family G-proteins to the function of thalamocortical relay (TC) neurons. Disruption of Galpha(q) function in a conditional forebrain-specific Galpha(q)/Galpha(11)-double-deficient mouse line (Galpha(q)/Galpha(11)(-/-) had no effects on the resting membrane potential (V (rest)) and the amplitude of the standing outward current (I (SO)). Stimulation of muscarinic acetylcholine (ACh) receptors (mAChR; muscarine, 50 microM) induced a decrease in I (SO) amplitude in wild-type mice (36 +/- 4%, n = 5), a constitutive Galpha(11)-deficient mouse line (Galpha(11)(-/-; 36 +/- 3%, n = 8), and Galpha(q)/Galpha(11)(-/-) (11 +/- 2%, n = 16). Current-clamp recordings revealed a muscarine-induced positive shift in V (rest) of 23 +/- 2 mV (n = 6), 18 +/- 5 mV (n = 5), and 2 +/- 1 mV (n = 9) in wild type, Galpha(11)(-/-), and Galpha(q)/Galpha(11)(-/-), respectively. This depolarization was associated with a change in TC neuron activity from burst to tonic firing in wild type and Galpha(11)(-/-), but not in Galpha(q)/Galpha(11)(-/-). The use of specific antibodies and of pharmacological agents with preferred affinity points to the contribution of m(1)AChR and m(3)AChR. In conclusion, we present two novel aspects of the physiology of the thalamocortical system by demonstrating that the depolarization of TC neurons, which is induced by the action of transmitters of ascending brainstem fibers, is governed roughly equally by both m(1)AChR and m(3)AChR and is transduced by Galpha(q) but not by Galpha(11).


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, Muscarinic/physiology , Thalamus/physiology , Animals , Blotting, Western , DNA Primers/chemistry , Electrophysiology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Immunohistochemistry , Integrases/metabolism , Integrases/physiology , Lac Operon/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/pharmacology , Neurons/physiology , Patch-Clamp Techniques , Receptors, Muscarinic/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Thalamus/drug effects , Thalamus/metabolism
18.
Dev Neurobiol ; 68(7): 934-49, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18361402

ABSTRACT

Networks of cortical neurons in vitro spontaneously develop synchronous oscillatory electrical activity at around the second week in culture. However, the underlying mechanisms and in particular the role of GABAergic interneurons in initiation and synchronization of oscillatory activity in developing cortical networks remain elusive. Here, we examined the intrinsic properties and the development of GABAergic and glutamatergic input onto presumed projection neurons (PNs) and large interneurons (L-INs) in cortical cultures of GAD67-GFP mice. Cultures developed spontaneous synchronous activity already at 5-7 days in vitro (DIV), as revealed by imaging transient changes in Fluo-3 fluorescence. Concurrently, spontaneous glutamate-mediated and GABA(A)-mediated postsynaptic currents (sPSCs) occured at 5 DIV. For both types of neurons the frequency of glutamatergic and GABAergic sPSCs increased with DIV, whereas the charge transfer of glutamatergic sPSCs increased and the charge transfer of GABAergic sPSCs decreased with cultivation time. The ratio between GABAergic and the overall charge transfer was significantly reduced with DIV for L-INs and PNs, indicating an overall reduction in GABAergic synaptic drive with maturation of the network. In contrast, analysis of miniature PSCs (mPSCs) revealed no significant changes of charge transfer with DIV for both types of neurons, indicating that the reduction in GABAergic drive was not due to a decreased number of functional synapses. Our data suggest that the global reduction in GABAergic synaptic drive together with more synaptic input to PNs and L-INs during maturation may enhance rhythmogenesis of the network and increase the synchronization at the level of population bursts.


Subject(s)
Down-Regulation/physiology , Gene Expression Regulation, Developmental/physiology , Neocortex/cytology , Nerve Net/physiology , Synapses/pathology , gamma-Aminobutyric Acid/metabolism , Animals , Bicuculline/pharmacology , Cells, Cultured , Dose-Response Relationship, Radiation , Electric Stimulation , Embryo, Mammalian , GABA Antagonists/pharmacology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Interneurons/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/embryology , Nerve Net/embryology , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Picrotoxin/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors
19.
Neuropharmacology ; 53(3): 431-46, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17675191

ABSTRACT

Channel blocking, anti-oscillatory, and anti-epileptic effects of clinically used anti-absence substances (ethosuximide, valproate) and the T-type Ca2+ current (IT) blocker mibefradil were tested by analyzing membrane currents in acutely isolated local circuit interneurons and thalamocortical relay (TC) neurons, slow intrathalamic oscillations in brain slices, and spike and wave discharges (SWDs) occurring in vivo in Wistar Albino Glaxo rats from Rijswijk (WAG/Rij). Substance effects in vitro were compared between WAG/Rij and a non-epileptic control strain, the ACI rats. Ethosuximide (ETX) and valproate were found to block IT in acutely isolated thalamic neurons. Block of IT by therapeutically relevant ETX concentrations (0.25-0.75 mM) was stronger in WAG/Rij, although the maximal effect at saturating concentrations (>or=10 mM) was stronger in ACI. Ethosuximide delayed the onset of the low threshold Ca2+ spike (LTS) of neurons recorded in slice preparations. Mibefradil (>or=2 microM) completely blocked IT and the LTS, dampened evoked thalamic oscillations, and attenuated SWDs in vivo. Computational modeling demonstrated that the complete effect of ETX can be replicated by a sole reduction of IT. However, the necessary degree of IT reduction was not induced by therapeutically relevant ETX concentrations. A combined reduction of IT, the persistent sodium current, and the Ca2+ activated K+ current resulted in an LTS alteration resembling the experimental observations. In summary, these results support the hypothesis of IT reduction as part of the mechanism of action of anti-absence drugs and demonstrate the ability of a specific IT antagonist to attenuate rhythmic burst firing and SWDs.


Subject(s)
Anticonvulsants/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Epilepsy, Absence/pathology , Interneurons/drug effects , Thalamus/pathology , Animals , Animals, Newborn , Anticonvulsants/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Electroencephalography , Epilepsy, Absence/drug therapy , Ethosuximide/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mibefradil/pharmacology , Patch-Clamp Techniques/methods , Rats , Rats, Inbred ACI
20.
Mol Cell Neurosci ; 35(2): 356-67, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17482476

ABSTRACT

Projection neurons of mouse basolateral amygdala responded to CCK with an inward current at a holding potential of -70 mV. This response was mediated by CCK2 receptors as indicated by agonist and antagonist effectiveness, and conveyed via G-proteins of the G(q/11) family as it was abolished in gene knockout mice. Maximal current amplitude was insensitive to extracellular potassium, cesium, and calcium ions, respectively, whereas amplitude and reversal potential critically depended upon extracellular sodium concentration. The current reversed near -20 mV consistent with activation of a mixed cationic channel reminiscent of transient receptor potential (TRP) channels. Extracellular application of the non-selective TRP channel blockers 2-APB, flufenamic acid, Gd3+, and ruthenium red, respectively, inhibited CCK induced inward currents. Single cell PCR confirmed the expression of TRPC1,4,5 and coexpression of TRPC1 with TRPC4 or TRPC5 in some cells. CCK responses were associated with depolarization leading to an increase in cell excitability.


Subject(s)
Amygdala/cytology , Cholecystokinin/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Neurons/drug effects , Synapses/drug effects , Transient Receptor Potential Channels/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Drug Interactions , Excitatory Postsynaptic Potentials/physiology , Female , GTP-Binding Protein alpha Subunits/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...