Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
NPJ Parkinsons Dis ; 10(1): 61, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491070

ABSTRACT

The synthetic tetrahydrocannabinol-analog nabilone improved non-motor symptoms (NMS) in Parkinson's disease (PD) patients in a placebo-controlled, double-blind, parallel-group, randomized withdrawal trial with enriched enrollment (NMS-Nab-study). This was a single-center open-label extension study to assess the long-term safety and efficacy of nabilone for NMS in PD. To be eligible for this study, patients had to be treatment responders during the previous NMS-Nab-trial and complete its double-blind phase without experiencing a drug-related serious/severe/moderate adverse event (AE). Patients were re-introduced to nabilone during an up-titration phase until their overall NMS burden improved. Nabilone was continued for six months with clinic visits every 3 months. Evaluation of AEs was based on self-report and clinical assessment. Twenty-two patients participated in the NMS-Nab2-study (age-median 68.33 y, 52% females, disease duration-median 7.42 y). Nabilone was well tolerated with concentration difficulties as the most common treatment-related AE (possibly/not related n = 1 each). One in two drop-outs discontinued because of an AE for which a prohibited concomitant medication needed to be introduced (night-time sleep problems). Efficacy evaluation showed a significant and lasting improvement in NMS burden according to the CGI-I (79% at V3). Nabilone improved overall sleep (NMSS Domain-2: -8.26 points; 95%CI -13.82 to -2.71; p = 0.004; ES = -0.72), night-time sleep problems (MDS-UPDRS-1.7: -1.42 points; 95 CI -2.16 to -0.68; p = 0.002; ES = -0.92), and overall pain (KPPS Total Score: -8.00 points; 95%CI -15.05 to -0.95; p = 0.046; ES -0.55 and MDS-UPDRS-1.9: -0.74 points; 95%CI -1.21 to -0.26; p = 0.008; ES = -0.74). This study demonstrates continuous long-term safety and efficacy in PD patients responding early to nabilone without intolerable side effects.

2.
Mov Disord Clin Pract ; 9(6): 751-758, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35937495

ABSTRACT

Background: The synthetic tetrahydrocannabinol analogue nabilone improved overall non-motor symptom (NMS) burden in Parkinson's disease (PD) patients in comparison to placebo. Objectives: To characterize the effects of nabilone on different sleep outcomes in PD patients. Methods: We performed a post-hoc analysis of the controlled, double-blind, enriched enrollment randomized withdrawal NMS-Nab study to assess the effects of nabilone on sleep outcomes in study participants who reported clinically-relevant sleep problems (MDS-UPDRS-1.7 ≥ 2 points). Results: After open-label nabilone administration, 77.4% reported no relevant sleep problem. In the withdrawal phase of the trial, the MDS-UPDRS-1.7. and the NMS-Scale Domain 2 (i.e., Sleep/Fatigue) significantly worsened only in PD patients in the placebo group, which was mostly driven by a significant worsening of insomnia (question 5 of the NMS-Scale Domain 2). Conclusions: This post-hoc analysis of the NMS-Nab trial suggests that nabilone has beneficial effects on sleep outcomes in PD patients experiencing sleep problems at baseline.The original trial was registered with ClinicalTrials.gov (NCT03769896, https://clinicaltrials.gov/ct2/show/NCT03769896) and EudraCT (2017-000192-86).

3.
Ann Neurol ; 88(4): 712-722, 2020 10.
Article in English | MEDLINE | ID: mdl-32757413

ABSTRACT

OBJECTIVE: The objective of this study was to assess the efficacy and safety of nabilone, a synthetic tetrahydrocannabinol analogue, as a treatment for non-motor symptoms (NMS) in Parkinson's disease (PD). METHODS: This was a phase II placebo-controlled, double-blind, parallel-group, enriched enrollment randomized withdrawal trial conducted at the Medical University Innsbruck. A random sample of 47 patients with PD with stable motor disease and disturbing NMS defined by a score of ≥4 points on the Movement Disorder Society - Unified PD Rating Scale-I (MDS-UPDRS-I) underwent open-label nabilone titration (0.25 mg once daily to 1 mg twice daily, phase I). Responders were randomized 1:1 to continue with nabilone or switch to placebo for 4 weeks (phase II). The primary efficacy criterion was the change of the MDS-UPDRS-I between randomization and week 4. Safety was analyzed in all patients who received at least one nabilone dose. RESULTS: Between October 2017 and July 2019, 19 patients received either nabilone (median dose = 0.75 mg) or placebo. At week 4, mean change of the MDS-UPDRS-I was 2.63 (95% confidence interval [CI] 1.53 to 3.74, p = 0.002, effect size = 1.15) in the placebo versus 1.00 (95% CI -0.16 to 2.16, p = 0.280, effect size = 0.42) in the nabilone-group (difference: 1.63, 95% CI 0.09 to 3.18, p = 0.030, effect size = 0.66). Seventy-seven percent of patients had adverse events (AEs) during open-label titration, most of them were transient. In the double-blind phase, similar proportions of patients in each group had AEs (42% in the placebo group and 32% in the nabilone group). There were no serious AEs. INTERPRETATION: Our results highlight the potential efficacy of nabilone for patients with PD with disturbing NMS, which appears to be driven by positive effects on anxious mood and night-time sleep problems. TRIAL REGISTRY: ClinicalTrials.gov (NCT03769896) and EudraCT (2017-000192-86). ANN NEUROL 2020;88:712-722.


Subject(s)
Dronabinol/analogs & derivatives , Parkinson Disease/drug therapy , Aged , Anxiety/etiology , Double-Blind Method , Dronabinol/therapeutic use , Female , Humans , Male , Middle Aged , Parkinson Disease/complications , Sleep Wake Disorders/etiology , Treatment Outcome
4.
J Neural Transm (Vienna) ; 126(8): 1061-1072, 2019 08.
Article in English | MEDLINE | ID: mdl-31129719

ABSTRACT

Although open-label observations report a positive effect of cannabinoids on non-motor symptoms (NMS) in Parkinson's disease (PD) patients, these effects remain to be investigated in a controlled trial for a broader use in NMS in PD patients. Therefore, we decided to design a proof-of-concept study to assess the synthetic cannabinoid nabilone for the treatment of NMS. We hypothesize that nabilone will improve NMS in patients with PD and have a favorable safety profile. The NMS-Nab Study is as a mono-centric phase II, randomized, placebo-controlled, double-blind, parallel-group, enriched enrollment withdrawal study. The primary efficacy criterion will be the change in Movement Disorders Society-Unified Parkinson's Disease-Rating Scale Part I score between baseline (i.e. randomization) and week 4. A total of 38 patients will have 80% power to detect a probability of 0.231 that an observation in the treatment group is less than an observation in the placebo group using a Wilcoxon rank-sum test with a 0.050 two-sided significance level assuming a true difference of 2.5 points between nabilone and placebo in the primary outcome measure and a standard deviation of the change of 2.4 points. The reduction of harm through an ineffective treatment, the possibility of individualized dosing, the reduction of sample size, and the possible evaluation of the influence of the placebo effect on efficacy outcomes justify this design for a single-centered placebo-controlled investigator-initiated trial of nabilone. This study should be the basis for further evaluations of long-term efficacy and safety of the use of cannabinoids in PD patients.


Subject(s)
Cannabinoid Receptor Agonists/therapeutic use , Dronabinol/analogs & derivatives , Parkinson Disease/drug therapy , Antiparkinson Agents/therapeutic use , Clinical Trial Protocols as Topic , Double-Blind Method , Dronabinol/therapeutic use , Female , Humans , Male , Patient Selection , Proof of Concept Study , Treatment Outcome
5.
Brain Struct Funct ; 223(6): 2663-2671, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29564531

ABSTRACT

In this study, we investigated the tissue expression levels, alpha subunit composition and distribution of Shaker-related voltage-dependent potassium Kv1 channels in human hippocampus by combining western blotting experiments, toxin autoradiography, in vivo radioligand binding studies, immunoprecipitation and immunohistochemistry. Tissue expression of Kv1.1 and Kv1.2 α-subunits in human post-mortem brain tissue was confirmed in immunoblot analysis using a panel of specific monoclonal and polyclonal antibodies. Immunoprecipitation experiments using toxin-prelabeled Kv1 channels revealed that all toxin-sensitive Kv1 channels in human hippocampus contained either a Kv1.1 or Kv1.2 α-subunit with the majority being composed of Kv1.1/Kv1.2 heterotetramers. Receptor autoradiography suggested Kv1.1/Kv1.2 channel expression in the molecular layer of dentate gyrus. In accordance, immunohistochemical experiments also observed Kv1.1 and Kv1.2 α-subunits in the molecular layer of the dentate gyrus, in addition to the CA3 stratum lucidum and the CA1 stratum oriens. These findings indicate expression in axons and terminals of hippocampal pathways, namely the perforant path, the mossy fiber pathway and the Schaffer collaterals. Herein we present the first direct demonstration that Kv1.1 and Kv1.2 channel proteins are targeted to distinct compartments of the human hippocampal formation and that this expression pattern largely reflects their distribution profile in murine brain.


Subject(s)
Hippocampus/metabolism , Shaker Superfamily of Potassium Channels/metabolism , Aged , Animals , Autoradiography , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Female , Hippocampus/cytology , Humans , Immunoprecipitation , Iodine Isotopes/pharmacokinetics , Kv1.2 Potassium Channel/metabolism , Male , Mice , Middle Aged , Scorpion Venoms/pharmacokinetics
6.
Brain Struct Funct ; 222(2): 973-979, 2017 03.
Article in English | MEDLINE | ID: mdl-27357310

ABSTRACT

SK2 (KCa2.2) channels are voltage-independent Ca2+-activated K+ channels that regulate neuronal excitability in brain regions important for memory formation. In this study, we investigated the distribution and expression of SK2 channels in human brain by Western blot analysis and immunohistochemistry. Immunoblot analysis of human brain indicated expression of four distinct SK2 channel isoforms: the standard, the long and two short isoforms. Immunohistochemistry in paraffin-embedded post-mortem brain sections was performed in the hippocampal formation, amygdala and neocortex. In hippocampus, SK2-like immunoreactivity could be detected in strata oriens and radiatum of area CA1-CA2 and in the molecular layer. In the amygdala, SK2-like immunoreactivity was highest in the basolateral nuclei, while in neocortex, staining was mainly found enriched in layer V. Activation of SK2 channels is thought to regulate neuronal excitability in brain by contributing to the medium afterhyperpolarization. However, SK2 channels are blocked by apamin with a sensitivity that suggests heteromeric channels. The herein first shown expression of SK2 human isoform b in brain could explain the variability of electrophysiological findings observed with SK2 channels.


Subject(s)
Brain/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Aged , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Protein Isoforms/metabolism , Small-Conductance Calcium-Activated Potassium Channels/immunology
7.
J Comp Neurol ; 524(10): 2093-116, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26587966

ABSTRACT

The sodium-activated potassium channels Slick (Slo2.1, KCNT2) and Slack (Slo2.2, KCNT1) are high-conductance potassium channels of the Slo family. In neurons, Slick and Slack channels are involved in the generation of slow afterhyperpolarization, in the regulation of firing patterns, and in setting and stabilizing the resting membrane potential. The distribution and subcellular localization of Slick and Slack channels in the mouse brain have not yet been established in detail. The present study addresses this issue through in situ hybridization and immunohistochemistry. Both channels were widely distributed and exhibited distinct distribution patterns. However, in some brain regions, their expression overlapped. Intense Slick channel immunoreactivity was observed in processes, varicosities, and neuronal cell bodies of the olfactory bulb, granular zones of cortical regions, hippocampus, amygdala, lateral septal nuclei, certain hypothalamic and midbrain nuclei, and several regions of the brainstem. The Slack channel showed primarily a diffuse immunostaining pattern, and labeling of cell somata and processes was observed only occasionally. The highest Slack channel expression was detected in the olfactory bulb, lateral septal nuclei, basal ganglia, and distinct areas of the midbrain, brainstem, and cerebellar cortex. In addition, comparing our data obtained from mouse brain with a previously published study on rat brain revealed some differences in the expression and distribution of Slick and Slack channels in these species. J. Comp. Neurol. 524:2093-2116, 2016. © 2015 The Authors The Journal of Comparative Neurology Published by Wiley Periodicals, Inc.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , RNA, Messenger/metabolism , Transfection
8.
Biochem Biophys Rep ; 4: 291-298, 2015 Dec.
Article in English | MEDLINE | ID: mdl-29124216

ABSTRACT

The sodium-activated potassium channels Slick (Slo2.1, KCNT2) and Slack (Slo2.2, KCNT1) are paralogous channels of the Slo family of high-conductance potassium channels. Slick and Slack channels are widely distributed in the mammalian CNS and they play a role in slow afterhyperpolarization, generation of depolarizing afterpotentials and in setting and stabilizing the resting potential. In the present study we used a combined approach of (co)-immunoprecipitation studies, Western blot analysis, double immunofluorescence and mass spectrometric sequencing in order to investigate protein-protein interactions of the Slick and Slack channels. The data strongly suggest that Slick and Slack channels co-assemble into identical cellular complexes. Double immunofluorescence experiments revealed that Slick and Slack channels co-localize in distinct mouse brain regions. Moreover, we identified the small cytoplasmic protein beta-synuclein and the transmembrane protein 263 (TMEM 263) as novel interaction partners of both, native Slick and Slack channels. In addition, the inactive dipeptidyl-peptidase (DPP 10) and the synapse associated protein 102 (SAP 102) were identified as constituents of the native Slick and Slack channel complexes in the mouse brain. This study presents new insights into protein-protein interactions of native Slick and Slack channels in the mouse brain.

9.
Biochim Biophys Acta ; 1833(12): 3166-3175, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036102

ABSTRACT

The Transient Receptor Potential Vanilloid 1 (TRPV1, vanilloid receptor 1) ion channel plays a key role in the perception of thermal and inflammatory pain, however, its molecular environment in dorsal root ganglia (DRG) is largely unexplored. Utilizing a panel of sequence-directed antibodies against TRPV1 protein and mouse DRG membranes, the channel complex from mouse DRG was detergent-solubilized, isolated by immunoprecipitation and subsequently analyzed by mass spectrometry. A number of potential TRPV1 interaction partners were identified, among them cytoskeletal proteins, signal transduction molecules, and established ion channel subunits. Based on stringent specificity criteria, the voltage-gated K(+) channel beta 2 subunit (Kvß2), an accessory subunit of voltage-gated K(+) channels, was identified of being associated with native TRPV1 channels. Reverse co-immunoprecipitation and antibody co-staining experiments confirmed TRPV1/Kvß2 association. Biotinylation assays in the presence of Kvß2 demonstrated increased cell surface expression levels of TRPV1, while patch-clamp experiments resulted in a significant increase of TRPV1 sensitivity to capsaicin. Our work shows, for the first time, the association of a Kvß subunit with TRPV1 channels, and suggests that such interaction may play a role in TRPV1 channel trafficking to the plasma membrane.


Subject(s)
Protein Subunits/metabolism , Shaker Superfamily of Potassium Channels/metabolism , TRPV Cation Channels/metabolism , Animals , Biotinylation , Cell Membrane/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Knockout , Patch-Clamp Techniques , Protein Binding , Rats , Recombinant Proteins/metabolism , Shaker Superfamily of Potassium Channels/chemistry
10.
Biochim Biophys Acta ; 1833(6): 1421-33, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23454728

ABSTRACT

RTN1A is a reticulon protein with predominant localization in the endoplasmic reticulum (ER). It was previously shown that RTN1A is expressed in neurons of the mammalian central nervous system but functional information remains sparse. To elucidate the neuronal function of RTN1A, we chose to focus our investigation on identifying possible novel binding partners specifically interacting with the unique N-terminus of RTN1A. Using a nonbiased approach involving GST pull-downs and MS analysis, we identified the intracellular calcium release channel ryanodine receptor 2 (RyR2) as a direct binding partner of RTN1A. The RyR2 binding site was localized to a highly conserved 150-amino acid residue region. RTN1A displays high preference for RyR2 binding in vitro and in vivo and both proteins colocalize in hippocampal neurons and Purkinje cells. Moreover, we demonstrate the precise subcellular localization of RTN1A in Purkinje cells and show that RTN1A inhibits RyR channels in [(3)H]ryanodine binding studies on brain synaptosomes. In a functional assay, RTN1A significantly reduced RyR2-mediated Ca(2+) oscillations. Thus, RTN1A and RyR2 might act as functional partners in the regulation of cytosolic Ca(2+) dynamics the in neurons.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Binding Sites , Blotting, Western , Cells, Cultured , Cytosol/metabolism , Hippocampus/cytology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Neurons/cytology , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ryanodine/metabolism , Tandem Mass Spectrometry
11.
Proc Natl Acad Sci U S A ; 107(34): 14950-7, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20668236

ABSTRACT

Local Ca(2+) signaling occurring within nanometers of voltage-gated Ca(2+) (Cav) channels is crucial for CNS function, yet the molecular composition of Cav channel nano-environments is largely unresolved. Here, we used a proteomic strategy combining knockout-controlled multiepitope affinity purifications with high-resolution quantitative MS for comprehensive analysis of the molecular nano-environments of the Cav2 channel family in the whole rodent brain. The analysis shows that Cav2 channels, composed of pore-forming alpha1 and auxiliary beta subunits, are embedded into protein networks that may be assembled from a pool of approximately 200 proteins with distinct abundance, stability of assembly, and preference for the three Cav2 subtypes. The majority of these proteins have not previously been linked to Cav channels; about two-thirds are dedicated to the control of intracellular Ca(2+) concentration, including G protein-coupled receptor-mediated signaling, to activity-dependent cytoskeleton remodeling or Ca(2+)-dependent effector systems that comprise a high portion of the priming and release machinery of synaptic vesicles. The identified protein networks reflect the cellular processes that can be initiated by Cav2 channel activity and define the molecular framework for organization and operation of local Ca(2+) signaling by Cav2 channels in the brain.


Subject(s)
Brain/metabolism , Calcium Channels/metabolism , Amino Acid Sequence , Animals , Calcium Channels/chemistry , Calcium Channels/deficiency , Calcium Channels/genetics , Calcium Signaling , In Vitro Techniques , Mice , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Protein Stability , Protein Subunits , Proteome , Proteomics/methods , Rats
12.
Brain Res ; 1348: 187-99, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20553876

ABSTRACT

Small conductance calcium (Ca(2+)) activated SK channels are critical regulators of neuronal excitability in hippocampus. Accordingly, these channels are thought to play a key role in controlling neuronal activity in acute models of epilepsy. In this study, we investigate the expression and function of SK channels in the pilocarpine model of mesial temporal lobe epilepsy. For this purpose, protein expression was assessed using western blotting assays and gene expression was analyzed using TaqMan-based probes and the quantitative real-time polymerase chain reaction (qPCR) comparative method delta-delta cycle threshold ( big up tri, open big up tri, openCT) in samples extracted from control and epileptic rats. In addition, the effect of SK channel antagonist UCL1684 and agonist NS309 on CA1 evoked population spikes was studied in hippocampal slices. Western blotting analysis showed a significant reduction in the expression of SK1 and SK2 channels at 10days following status epilepticus (SE), but levels recovered at 1month and at more than 2months after SE. In contrast, a significant down-regulation of SK3 channels was detected after 10days of SE. Analysis of gene expression by qPCR revealed a significant reduction of transcripts for SK2 (Kcnn1) and SK3 (Kcnn3) channels as early as 10days following pilocarpine-induced SE and during the chronic phase of the pilocarpine model. Moreover, bath application of UCL1684 (100nM for 15min) induced a significant increase of the population spike amplitude and number of spikes in the hippocampal CA1 area of slices obtained control and chronic epileptic rats. This effect was obliterated by co-administration of UCL1684 with SK channel agonist NS309 (1microM). Application of NS309 failed to modify population spikes in the CA1 area of slices taken from control and epileptic rats. These data indicate an abnormal expression of SK channels and a possible dysfunction of these channels in experimental MTLE.


Subject(s)
Gene Expression Regulation/drug effects , Membrane Potentials/drug effects , Muscarinic Agonists/adverse effects , Pilocarpine/adverse effects , Small-Conductance Calcium-Activated Potassium Channels/physiology , Status Epilepticus , Age Factors , Alkanes/pharmacology , Analysis of Variance , Animals , Disease Models, Animal , Drug Interactions , Hippocampus/pathology , In Vitro Techniques , Indoles/pharmacology , Male , Membrane Potentials/physiology , Neurons/drug effects , Neurons/physiology , Oximes/pharmacology , Quinolinium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/physiopathology , Time Factors
13.
J Alzheimers Dis ; 20(4): 1167-80, 2010.
Article in English | MEDLINE | ID: mdl-20413896

ABSTRACT

Cumulative evidence indicates that amyloid-beta peptides exert some of their neurodegenerative effects through modulation of L-type voltage gated calcium channels, which play key roles in a diverse range of CNS functions. In this study we examined the expression of CaV1.2 L-type voltage gated calcium channels in transgenic mice overexpressing human AbetaPP751 with the London (V717I) and Swedish (K670M/N671L) mutations by immunohistochemistry in light and electron microscopy. In hippocampal layers of wild type and transgenic mice, CaV1.2 channels were predominantly localized to somato-dendritic domains of neurons, and to astrocytic profiles with an age-dependent increase in labeling density. In transgenic animals, CaV1.2-like immunoreactive clusters were found in neuronal profiles in association with amyloid-beta plaques. Both the number and density of these clusters depended upon age of animals and number of plaques. The most striking difference between wild type and transgenic mice was the age-dependent expression of CaV1.2 channels in reactive astrocytes. At the age of 6 month, CaV1.2 channels were rarely detected in reactive astrocytes of transgenic mice, but an incremental number of CaV1.2 expressing reactive astrocytes was found with increasing age of animals and number of amyloid-beta plaques. This study demonstrates that CaV1.2 channels are highly expressed in reactive astrocytes of 12-months of age transgenic mice, which might be a consequence of the increasing amyloid burden. Further studies should clarify which functional implications are associated with the higher availability of CaV1.2 channels in late stage Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Aging/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/ultrastructure , Brain/pathology , Brain/ultrastructure , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Mutation , Neurites/drug effects , Neurites/ultrastructure
14.
J Comp Neurol ; 515(2): 215-30, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19412945

ABSTRACT

Calcium-activated potassium channels have been shown to be critically involved in neuronal function, but an elucidation of their detailed roles awaits identification of the microdomains where they are located. This study was undertaken to unravel the precise subcellular distribution of the large-conductance calcium-activated potassium channels (called BK, KCa1.1, or Slo1) in the somatodendritic compartment of cerebellar Purkinje cells by means of postembedding immunogold cytochemistry and SDS-digested freeze-fracture replica labeling (SDS-FRL). We found BK channels to be unevenly distributed over the Purkinje cell plasma membrane. At distal dendritic compartments, BK channels were scattered over the plasma membrane of dendritic shafts and spines but absent from postsynaptic densities. At the soma and proximal dendrites, BK channels formed two distinct pools. One pool was scattered over the plasma membrane, whereas the other pool was clustered in plasma membrane domains overlying subsurface cisterns. The labeling density ratio of clustered to scattered channels was about 60:1, established in SDS-FRL. Subsurface cisterns, also called hypolemmal cisterns, are subcompartments of the endoplasmic reticulum likely representing calciosomes that unload and refill Ca2+ independently. Purkinje cell subsurface cisterns are enriched in inositol 1,4,5-triphosphate receptors that mediate the effects of several neurotransmitters, hormones, and growth factors by releasing Ca2+ into the cytosol, generating local Ca2+ sparks. Such increases in cytosolic [Ca2+] may be sufficient for BK channel activation. Clustered BK channels in the plasma membrane may thus participate in building a functional unit (plasmerosome) with the underlying calciosome that contributes significantly to local signaling in Purkinje cells.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Purkinje Cells/physiology , Animals , Cell Membrane/physiology , Cell Membrane/ultrastructure , Dendritic Cells/metabolism , Excitatory Postsynaptic Potentials/drug effects , Freeze Fracturing , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Inositol 1,4,5-Trisphosphate Receptors/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Male , Mice , Mice, Inbred C57BL , Purkinje Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Sodium Dodecyl Sulfate , Tissue Embedding , gamma-Aminobutyric Acid/physiology
15.
J Alzheimers Dis ; 13(2): 123-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18376054

ABSTRACT

Chromogranin B and secretogranin II are major soluble constituents of large dense core vesicles of presynaptic structures and have been found in neuritic plaques of Alzheimer patients. We examined the distribution and expression of these peptides in both transgenic mice over expressing human amyloid-beta protein precursor APP751 with the London (V717I) and Swedish (K670M/N671L) mutations and in human post-mortem brain. In transgenic mice, the number of amyloid-beta plaques and chromogranin immunopositive plaques increased from 6 to 12 months. About 60% of amyloid-beta plaques were associated with chromogranin B and about 40% with secretogranin II. Chromogranin immunoreactivity appeared mainly as swollen dystrophic neurites. Neither synaptophysin- nor glial fibrillary acidic protein- immunoreactivity was expressed in chromogranin immunoreactive structures at any timepoint. Density of chromogranin peptides in hippocampal structures did not change in transgenic animals at any timepoint, even though animals had a poorer performance in the Morris water maze task. In conclusion, our findings in transgenic animals partly resembled findings in Alzheimer patients. Chromogranin peptides were associated with amyloid-beta plaques, but were not reduced in specific brain areas as previously reported by our group. Therefore specific changes of chromogranin peptides observed in Alzheimer patients can be related to amyloid-beta pathology only.


Subject(s)
Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Chromogranin B/genetics , Point Mutation/genetics , Secretogranin II/genetics , Animals , Aspartic Acid/genetics , Brain/metabolism , Brain/pathology , Chromogranin B/metabolism , Gene Expression/genetics , Glial Fibrillary Acidic Protein/metabolism , Isoleucine/genetics , Leucine/genetics , Methionine/genetics , Mice , Mice, Transgenic , Secretogranin II/metabolism , Substance P/genetics , Substance P/metabolism , Valine/genetics
16.
Brain Pathol ; 17(4): 389-98, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17784877

ABSTRACT

The latent persistence of herpes simplex virus type 1 (HSV-1) in human trigeminal ganglia (TG) is accompanied by a chronic CD8 T-cell infiltrate. The focus of the current work was to look for HSV-1 transcription activity as a potential trigger of the immune response and to characterize the immune cell infiltrates by this feature. We combined in situ hybridization, laser cutting microscopy, and single cell RT-PCR to demonstrate the expression of the HSV-1 immediate early (IE) genes ICP0 and ICP4 in human trigeminal neurons. Using CDR3 spectratyping, we showed that the infiltrating T-cells are clonally expanded, indicating an antigen-driven immune response. Moreover, the persisting CD8+ T-cells had features of the memory effector phenotype. The voltage-gated potassium channel Kv1.3, a marker of chronic activated memory effector cells, and the chemokines CCL5 and CXCL10 were expressed by a subpopulation of infiltrating cells. The corresponding chemokine receptors CCR5 and CXCR3 were co-expressed on virtually all CD8 T-cells. In addition, T-cells expressed granzymes and perforin. In contrast to animal models of HSV-1 latency, hardly any FoxP3-positive regulatory T-cells were detected in human TG. Thus, HSV-1 IE genes are expressed in human TG and the infiltrating T-cells bear several characteristics that suggest viral antigenic stimulation.


Subject(s)
Genes, Immediate-Early/genetics , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , T-Lymphocytes/virology , Trigeminal Ganglion/virology , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chemokines/immunology , Chemokines/metabolism , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Clone Cells/immunology , Clone Cells/virology , Female , Gene Expression Regulation, Viral/genetics , Genes, Viral/genetics , Herpes Simplex/physiopathology , Herpesvirus 1, Human/immunology , Humans , Immunologic Memory/genetics , Immunologic Memory/immunology , Kv1.3 Potassium Channel/metabolism , Male , Middle Aged , Neurons, Afferent/immunology , Neurons, Afferent/virology , Phenotype , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology , Trigeminal Ganglion/cytology , Trigeminal Ganglion/immunology , Virus Latency/genetics , Virus Latency/immunology
17.
Brain Res ; 1143: 199-207, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17328871

ABSTRACT

Substance P-like immunoreactivity (-LI) is found in neuritic plaques, and is reduced in patients suffering from Alzheimer disease (AD). In this study, we examined the distribution and expression of substance P in transgenic mice overexpressing human amyloid precursor protein (hAPP) APP751 with the London (V717I) and Swedish (K670M/N671L) mutations. Immunohistochemistry was performed to localize substance P- and glial fibrillary acidic protein-LI by confocal microscopy. In hAPP transgenic mice, the number of beta-amyloid plaques significantly increased from 6 to 12 months. About 5% of beta-amyloid plaques were substance P-immunoreactive. In transgenic mice, the morphology of substance P-immunoreactive structures changed by consisting of swollen and dystrophic neurites mostly associated with beta-amyloid plaques. The overall localization and the relative substance P densities were not different between wild type and transgenic mice at 6 and 12 months. At month 12, a dramatic change in the distribution pattern of substance P-LI was observed as it was now expressed in a high number of reactive astrocytes. This expression of substance P in astrocytes was mainly found in the hippocampal formation and thalamic nuclei with a preferential association with beta-amyloid plaques, whereas in cortical regions only faintly substance P-immunoreactive astrocytes were observed. This study indicates that substance P undergoes complex changes in this animal Alzheimer disease model. Future experiments including substance P antagonists are necessary to further explore the interaction between beta-amyloid deposits and substance P.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Gene Expression/genetics , Mutation , Substance P/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Isoleucine/genetics , Leucine/genetics , Lysine/genetics , Methionine/genetics , Mice , Mice, Transgenic , Substance P/genetics , Valine/genetics
18.
Science ; 314(5799): 615-20, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17068255

ABSTRACT

Large-conductance calcium- and voltage-activated potassium channels (BKCa) are dually activated by membrane depolarization and elevation of cytosolic calcium ions (Ca2+). Under normal cellular conditions, BKCa channel activation requires Ca2+ concentrations that typically occur in close proximity to Ca2+ sources. We show that BKCa channels affinity-purified from rat brain are assembled into macromolecular complexes with the voltage-gated calcium channels Cav1.2 (L-type), Cav2.1 (P/Q-type), and Cav2.2 (N-type). Heterologously expressed BKCa-Cav complexes reconstitute a functional "Ca2+ nanodomain" where Ca2+ influx through the Cav channel activates BKCa in the physiological voltage range with submillisecond kinetics. Complex formation with distinct Cav channels enables BKCa-mediated membrane hyperpolarization that controls neuronal firing pattern and release of hormones and transmitters in the central nervous system.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Calcium/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Brain Chemistry , CHO Cells , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/isolation & purification , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/isolation & purification , Calcium Signaling , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Cricetinae , Cricetulus , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/isolation & purification , Membrane Potentials/drug effects , Molecular Sequence Data , Patch-Clamp Techniques , Rats , Transfection , Xenopus
19.
Eur J Neurosci ; 24(2): 442-54, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16903852

ABSTRACT

Neurons are highly specialized cells in which the integration and processing of electrical signals critically depends on the precise localization of ion channels. For large-conductance Ca(2+)- activated K(+) (BK) channels, targeting to presynaptic membranes in hippocampal pyramidal cells was reported; however, functional evidence also suggests a somatodendritic localization. Therefore we re-examined the subcellular distribution of BK channels in mouse hippocampus using a panel of independent antibodies in a combined approach of conventional immunocytochemistry on cultured neurons, pre- and postembedding electron microscopy and immunoprecipitation. In cultured murine hippocampal neurons, the colocalization of BK channels with both pre- and postsynaptic marker proteins was observed. Electron microscopy confirmed targeting of BK channels to axonal as well as dendritic membranes of glutamatergic synapses in hippocampus. A postsynaptic localization of BK channels was also supported by the finding that the channel coimmunoprecipitated with PSD95, a protein solely expressed in the postsynaptic compartment. These results thus demonstrate that BK channels reside in both post- and presynaptic compartments of hippocampal pyramidal neurons.


Subject(s)
Dendrites/metabolism , Hippocampus/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Presynaptic Terminals/metabolism , Pyramidal Cells/metabolism , Synaptic Membranes/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , Dendrites/ultrastructure , Disks Large Homolog 4 Protein , Guanylate Kinases , Hippocampus/ultrastructure , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Presynaptic Terminals/ultrastructure , Protein Subunits/metabolism , Pyramidal Cells/ultrastructure , Receptors, Glutamate/metabolism , Synaptic Membranes/ultrastructure , Synaptic Transmission/physiology
20.
Neuron ; 49(5): 697-706, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16504945

ABSTRACT

The voltage-gated potassium (Kv) channel subunit Kv1.1 is a major constituent of presynaptic A-type channels that modulate synaptic transmission in CNS neurons. Here, we show that Kv1.1-containing channels are complexed with Lgi1, the functionally unassigned product of the leucine-rich glioma inactivated gene 1 (LGI1), which is causative for an autosomal dominant form of lateral temporal lobe epilepsy (ADLTE). In the hippocampal formation, both Kv1.1 and Lgi1 are coassembled with Kv1.4 and Kvbeta1 in axonal terminals. In A-type channels composed of these subunits, Lgi1 selectively prevents N-type inactivation mediated by the Kvbeta1 subunit. In contrast, defective Lgi1 molecules identified in ADLTE patients fail to exert this effect resulting in channels with rapid inactivation kinetics. The results establish Lgi1 as a novel subunit of Kv1.1-associated protein complexes and suggest that changes in inactivation gating of presynaptic A-type channels may promote epileptic activity.


Subject(s)
Brain/metabolism , Kv1.1 Potassium Channel/physiology , Kv1.2 Potassium Channel/physiology , Neural Inhibition/physiology , Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western/methods , Brain/cytology , Brain Chemistry , Cell Membrane/metabolism , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Humans , Immunohistochemistry/methods , Intracellular Signaling Peptides and Proteins , Mass Spectrometry/methods , Membrane Potentials/physiology , Mutagenesis/physiology , Mutation , Oocytes , Patch-Clamp Techniques/methods , Protein Conformation , Rats , Sequence Alignment , Silver Staining/methods , Transfection/methods , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...