Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928162

ABSTRACT

Polyamine (PA) spermidine (SPD) plays a crucial role in aging. Since SPD accumulates in glial cells, particularly in Müller retinal cells (MCs), the expression of the SPD-synthesizing enzyme spermidine synthase (SpdS) in Müller glia and age-dependent SpdS activity are not known. We used immunocytochemistry, Western blot (WB), and image analysis on rat retinae at postnatal days 3, 21, and 120. The anti-glutamine synthetase (GS) antibody was used to identify glial cells. In the neonatal retina (postnatal day 3 (P3)), SpdS was expressed in almost all progenitor cells in the neuroblast. However, by day 21 (P21), the SpdS label was pronouncedly expressed in multiple neurons, while GS labels were observed only in radial Müller glial cells. During early cell adulthood, at postnatal day 120 (P120), SpdS was observed solely in ganglion cells and a few other neurons. Western blot and semi-quantitative analyses of SpdS labeling showed a dramatic decrease in SpdS at P21 and P120 compared to P3. In conclusion, the redistribution of SpdS with aging indicates that SPD is first synthesized in all progenitor cells and then later in neurons, but not in glia. However, MCs take up and accumulate SPD, regardless of the age-associated decrease in SPD synthesis in neurons.


Subject(s)
Ependymoglial Cells , Retina , Spermidine Synthase , Animals , Rats , Spermidine Synthase/metabolism , Spermidine Synthase/genetics , Retina/metabolism , Ependymoglial Cells/metabolism , Aging/metabolism , Spermidine/metabolism , Neuroglia/metabolism , Animals, Newborn
2.
Mol Psychiatry ; 27(4): 2315-2328, 2022 04.
Article in English | MEDLINE | ID: mdl-33190145

ABSTRACT

The striatum is the main input structure of the basal ganglia. Distinct striatal subfields are involved in voluntary movement generation and cognitive and emotional tasks, but little is known about the morphological and molecular differences of striatal subregions. The ventrolateral subfield of the striatum (VLS) is the orofacial projection field of the sensorimotor cortex and is involved in the development of orofacial dyskinesias, involuntary chewing-like movements that often accompany long-term neuroleptic treatment. The biological basis for this particular vulnerability of the VLS is not known. Potassium channels are known to be strategically localized within the striatum. In search of possible molecular correlates of the specific vulnerability of the VLS, we analyzed the expression of voltage-gated potassium channels in rodent and primate brains using qPCR, in situ hybridization, and immunocytochemical single and double staining. Here we describe a novel, giant, non-cholinergic interneuron within the VLS. This neuron coexpresses the vesicular GABA transporter, the calcium-binding protein parvalbumin (PV), and the Kv3.3 potassium channel subunit. This novel neuron is much larger than PV neurons in other striatal regions, displays characteristic electrophysiological properties, and, most importantly, is restricted to the VLS. Consequently, the giant striatal Kv3.3-expressing PV neuron may link compromised Kv3 channel function and VLS-based orofacial dyskinesias.


Subject(s)
Dyskinesias , Parvalbumins , Animals , Corpus Striatum/metabolism , Dyskinesias/metabolism , Interneurons/metabolism , Parvalbumins/metabolism , Potassium Channels/metabolism , Shaw Potassium Channels/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins
3.
Amino Acids ; 52(8): 1169-1180, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32816168

ABSTRACT

Polyamines (PAs), such as spermidine (SPD) and spermine (SPM), are essential to promote cell growth, survival, proliferation, and longevity. In the adult central nervous system (CNS), SPD and SPM are accumulated predominantly in healthy adult glial cells where PA synthesis is not present. To date, the accumulation and biosynthesis of PAs in developing astrocytes are not well understood. The purpose of the present study was to determine the contribution of uptake and/or synthesis of PAs using proliferation of neonatal astrocytes as an endpoint. We inhibited synthesis of PAs using α-difluoromethylornithine (DFMO; an inhibitor of the PA biosynthetic enzyme ornithine decarboxylase (ODC)) and inhibited uptake of PAs using trimer44NMe (PTI; a novel polyamine transport inhibitor). DFMO, but not PTI alone, blocked proliferation, suggesting that PA biosynthesis was present. Furthermore, exogenous administration of SPD rescued cell proliferation when PA synthesis was blocked by DFMO. When both synthesis and uptake of PAs were inhibited (DFMO + PTI), exogenous SPD no longer supported proliferation. These data indicate that neonatal astrocytes synthesize sufficient quantities of PAs de novo to support cell proliferation, but are also able to import exogenous PAs. This suggests that the PA uptake mechanism is present in both neonates as well as in adults and can support cell proliferation in neonatal astrocytes when ODC is blocked.


Subject(s)
Astrocytes/metabolism , Polyamines/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Eflornithine , Polyamines/antagonists & inhibitors , Protein Transport , Rats , Rats, Sprague-Dawley , Spermidine/metabolism , Spermine/metabolism
4.
Glia ; 66(10): 2200-2208, 2018 10.
Article in English | MEDLINE | ID: mdl-30151916

ABSTRACT

For long times astrocytes had been regarded as supporting cells, passively filling the spaces between neuronal cell bodies and their extensions. Now it is known that astrocytes are actively involved in a variety of important biological functions such as regulating cerebral blood flow, supporting neuronal metabolism, controlling the extracellular potassium concentration, and clearing neurotransmitters from the extracellular space. In line with this multitude of tasks astrocytes display conspicuous functional and regional heterogeneity. Using three complementary labeling methods nine classes of astrocytes have been differentiated, which were termed protoplasmic, fibrous, velate, radial, and perivascular astrocytes in addition to Bergmann, marginal, and ependymal glial cells. To complete this list retinal Müller cells and a largely forgotten astrocytic cell type, the "feathered cell" of Fañanas need to be added. So far, Fañanas cells could be only recognized with the tedious gold-sublimate procedure. Consequently, data indicating a potential biological function are completely missing. In a parallel investigation we used a battery of antibodies against potassium channels and related proteins to identify potential marker proteins for the immunocytochemical visualization of distinct cell types in the cerebellar cortex. Here we present novel marker proteins, the Kv2.2 potassium channel and calsenilin, to visualize Fañanas cells in the cerebellar Purkinje cell layer. Such markers will allow to identify Fañanas cell subsequent to patching and electrophysiological characterization. This may pave the path to obtain new functional data, which may be helpful to understand the role of these enigmatic cells in normal biological function and disease.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Fluorescent Antibody Technique , Neuroglia/cytology , Neuroglia/metabolism , Animals , Antibodies , Fluorescent Antibody Technique/methods , Gene Expression , Kv Channel-Interacting Proteins/metabolism , Male , Microscopy, Confocal , Rats, Wistar , Shab Potassium Channels/metabolism , Staining and Labeling
5.
Cell Mol Neurobiol ; 34(1): 61-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24077856

ABSTRACT

Among all K2P channels, TASK-3 shows the most widespread expression in rat brain, regulating neuronal excitability and transmitter release. Using a recently purified and characterized polyclonal monospecific antibody against TASK-3, the entire rat brain was immunocytochemically analyzed for expression of TASK-3 protein. Besides its well-known strong expression in motoneurons and monoaminergic and cholinergic neurons, TASK-3 expression was found in most neurons throughout the brain. However, it was not detected in certain neuronal populations, and neuropil staining was restricted to few areas. Also, it was absent in adult glial cells. In hypothalamic areas, TASK-3 was particularly strongly expressed in the supraoptic and suprachiasmatic nuclei, whereas other hypothalamic nuclei showed lower protein levels. Immunostaining of hippocampal CA1 and CA3 pyramidal neurons showed strongest expression, together with clear staining of CA3 mossy fibers and marked staining also in the dentate gyrus granule cells. In neocortical areas, most neurons expressed TASK-3 with a somatodendritic localization, most obvious in layer V pyramidal neurons. In the cerebellum, TASK-3 protein was found mainly in neurons and neuropil of the granular cell layer, whereas Purkinje cells were only faintly positive. Particularly weak expression was demonstrated in the forebrain. This report provides a comprehensive overview of TASK-3 protein expression in the rat brain.


Subject(s)
Brain Chemistry , Brain/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Brain/cytology , Immunohistochemistry , Male , Potassium Channels, Tandem Pore Domain/analysis , Protein Transport , Rats , Rats, Wistar
6.
PLoS One ; 19(7): e0304658, 2024.
Article in English | MEDLINE | ID: mdl-39052628

ABSTRACT

Polyamines (PAs) including putrescine (PUT), spermidine (SPD) and spermine (SPM) are small, versatile molecules with two or more positively charged amino groups. Despite their importance for almost all forms of life, their specific roles in molecular and cellular biology remain partly unknown. The molecular structures of PAs suggest two presumable biological functions: (i) as potential buffer systems and (ii) as interactants with poly-negatively charged molecules like nucleic acids. The present report focuses on the question, whether the molecular structures of PAs are essential for such functions, or whether other simple molecules like small peptides with closely spaced positively charged side chains might be suitable as well. Consequently, we created titration curves for PUT, SPD, and SPM, as well as for oligolysines like tri-, tetra-, and penta-lysine. None of the molecules provided substantial buffering capacity at physiological intracellular pH values. Apparently, the most important mechanism for intracellular pH homeostasis in neurons is not a buffer system but is provided by the actions of the sodium-hydrogen and the bicarbonate-chloride antiporters. In a similar approach we investigated the interaction with DNA by following the extinction at 260 nm when titrating DNA with the above molecules. Again, PUT and tri-lysine were not able to interact with herring sperm DNA, while SPD and SPM were. Obviously, the presence of several positively charged groups on its own is not sufficient for the interaction with nucleic acids. Instead, the precise spacing of these groups is necessary for biological activity.


Subject(s)
DNA , Peptides , Polyamines , RNA , Hydrogen-Ion Concentration , DNA/chemistry , DNA/metabolism , Buffers , RNA/chemistry , RNA/metabolism , Polyamines/chemistry , Polyamines/metabolism , Peptides/chemistry , Peptides/metabolism , Animals , Spermidine/chemistry , Spermidine/metabolism , Spermine/chemistry , Spermine/metabolism , Putrescine/chemistry , Putrescine/metabolism
7.
Neurobiol Dis ; 56: 14-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583611

ABSTRACT

In models of temporal lobe epilepsy, in-vitro exposure of the entorhinal cortex (EC) to low concentrations of acetylcholine (ACh) induces muscarinic-dependent seizure-like events. Potassium channels from the KCNQ/Kv7 family, which close upon activation of muscarinic receptors, are mutated in several epileptic syndromes such as benign familial neonatal convulsions (KCNQ2/KCNQ3) and sudden unexplained death in epilepsy (KCNQ1). Therefore, we tested the hypothesis whether the ictogenic effect of ACh involves alterations of KCNQ channels. In horizontal temporo-hippocampal slices from pilocarpine-treated chronically epileptic rats, field potential recordings of epileptiform activity were performed in response to the application of ACh, the KCNQ blocker linopirdine, and KCNQ agonists. In the EC of control rats, ACh (20 and 50 µM) induced nested fast activity in the range of 15-20 Hz riding on <1 Hz slow oscillations. By contrast, in slices from pilocarpine-treated rats, 5 µM ACh was sufficient to induce interictal discharges that frequently transformed to epileptiform events at 20 µM ACh. While the non-specific KCNQ/Kv7 channel blocker linopirdine (20 and 50 µM) had no effect in control animals, in slices from epileptic rats it induced interictal discharges or seizure-like events. These could be blocked by the unspecific KCNQ/Kv7 agonist retigabine and attenuated by the Kv7.1 agonist L364-373. Immunohistochemistry revealed reduced expression of KCNQ2 and KCNQ3 in the EC and of KCNQ3-positive dendrites in the subiculum of epileptic rats. These results indicate that channels of the KCNQ family are key regulators of seizure susceptibility and their decreased availability in the epileptic tissue may reduce seizure threshold and contribute to ictogenesis.


Subject(s)
Acetylcholine/pharmacology , Entorhinal Cortex/drug effects , KCNQ Potassium Channels/drug effects , Muscarinic Agonists/pharmacology , Pilocarpine/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Evoked Potentials/drug effects , Immunohistochemistry , Indoles/pharmacology , Male , Nerve Net/cytology , Nerve Net/physiology , Potassium Channel Blockers/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Seizures/physiopathology , Status Epilepticus/physiopathology
8.
Cereb Cortex ; 22(9): 2087-101, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22038909

ABSTRACT

Granule cells in the dentate gyrus are only sparsely active in vivo and survive hippocampal sclerosis (HS) during temporal lobe epilepsy better than neighboring cells. This phenomenon could be related to intrinsic properties specifically adapted to counteract excitation. We studied the mechanisms underlying the excitability of human granule cells using acute hippocampal slices obtained during epilepsy surgery. Patch-clamp recordings were combined with pharmacology, immunocytochemistry, and computer simulations. The input resistance of granule cells correlated negatively with the duration of epilepsy and the degree of HS. Hyperpolarization-activated, ZD7288-sensitive cation (I(H), HCN) currents and highly Ba(2+)-sensitive, inwardly rectifying K(+) (Kir) currents (and HCN1 and Kir2.2 protein) were present somatodendritically and further enhanced in patients with severe HS versus mild HS. The properties and function of I(H) were characterized in granule cells. Although I(H) depolarized the membrane, it strongly reduced the input resistance and shifted the current-frequency function to higher input values. The shunting influence of HCN and Kir was similar and these conductances correlated. Resonance was not observed. Simulations suggest that the combined upregulation of Kir and HCN conductances attenuates excitatory synaptic input, while stabilizing the membrane potential and responsiveness. Thus, granule cells homeostatically downscale their input-output transfer function during epilepsy.


Subject(s)
Adaptation, Physiological/physiology , Dentate Gyrus/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Epilepsy, Temporal Lobe/physiopathology , Humans , Organ Culture Techniques , Patch-Clamp Techniques
9.
J Neurol ; 270(12): 5866-5877, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37603074

ABSTRACT

Autoimmune encephalitides are seldom diseases. How rare they actually are, however, is not known. The low incidence combined with the problematic identification may dampen efforts of neurologists, to identify patients with unclear symptoms as suffering from autoimmune encephalitis. Here, we aim to obtain a better estimate, how many patients with autoimmune disorders should be expected among 100 inpatients in a conventional neurological department. From a total number of 2603 non-stroke patients attended in a 2-year period (2018-2019) 460 CSFs were obtained. From this collection 187 samples (40.7%, > 500 sections) could be analyzed with our immunocytochemical technique. Autoreactive antibodies were detected in 102 (55%) of these 187 CSF samples. Certainly, the presence of autoreactive antibodies does not necessarily indicate that the patient suffers from an autoimmune disease. Our data indicate that from roughly 2000 patients during 1 year about 125 patients with autoreactive CSF antibodies should be expected in a conventional neurological department. This represents the about 35-fold value of what is generally expected at present. Being aware of this high incidence may intensify the efforts of neurologist to identify patients with any type of autoimmune encephalitis. This will be beneficial for patients, because they often profit from immunomodulatory therapy. Interestingly, some CFSs from our patients react with the CA2 subdivision of the hippocampus. While long neglected, recent research places this area into an important position to influence hippocampal network physiology. Autoreactive antibodies in the CSF may disturb the function of CA2 neurons, thereby explaining some neuropsychiatric symptoms in patients with autoimmune encephalitides.


Subject(s)
Autoimmune Diseases of the Nervous System , Autoimmune Diseases , Encephalitis , Humans , Autoantibodies
10.
Biomolecules ; 13(7)2023 07 13.
Article in English | MEDLINE | ID: mdl-37509150

ABSTRACT

The importance of polyamines (PAs) for the central nervous system (CNS) is well known. Less clear, however, is where PAs in the brain are derived from. Principally, there are three possibilities: (i) intake by nutrition, release into the bloodstream, and subsequent uptake from CNS capillaries, (ii) production by parenchymatous organs, such as the liver, and again uptake from CNS capillaries, and (iii) uptake of precursors, such as arginine, from the blood and subsequent local biosynthesis of PAs within the CNS. The present investigation aimed to unequivocally answer the question of whether PAs, especially the higher ones like spermidine (SPD) and spermine (SPM), can or cannot be taken up into the brain from the bloodstream. For this purpose, a biotin-labelled analogue of spermine (B-X-SPM) was synthesized, characterized, and used to visualize its uptake into brain cells following application to acute brain slices, to the intraventricular space, or to the bloodstream. In acute brain slices there is strong uptake of B-X-SPM into protoplasmic and none in fibrous-type astrocytes. It is also taken up by neurons but to a lesser degree. Under in vivo conditions, astrocyte uptake of B-X-SPM from the brain interstitial fluid is also intense after intraventricular application. In contrast, following intracardial injection, there is no uptake from the bloodstream, indicating that the brain is completely dependent on the local synthesis of polyamines.


Subject(s)
Polyamines , Spermine , Spermidine , Brain , Neurons
11.
Cell Mol Neurobiol ; 32(2): 309-18, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22011781

ABSTRACT

Motor neurons are large cholinergic neurons located in the brain stem and spinal cord. In recent years, a functional role for TASK channels in cellular excitability and vulnerability to anesthetics of motor neurons has been described. Using a polyclonal monospecific antibody against the tandem pore domain K(+) channel (K2P channel) TWIK-related acid-sensitive K(+) channel (TASK-3), we analyzed the expression of the TASK-3 protein in motor systems of the rat CNS. Immunocytochemical staining showed strong TASK-3 expression in motor neurons of the facial, trigeminal, ambiguus, and hypoglossal nuclei. Oculomotor nuclei (including trochlear and abducens nucleus) were also strongly positive for TASK-3. The parasympathetic Edinger-Westphal nucleus and dorsal vagal nucleus showed significant, but weaker expression compared with somato- and branchiomotoric neurons. In addition, motor neurons in the anterior horn of the spinal cord were also strongly labeled for TASK-3 immunoreactivity. Based on morphological criteria, TASK-3 was found in the somatodendritic compartment of motor neurons. Cellular staining using methyl green and immunofluorescence double-labeling with anti-vesicular acetylcholine transporter (anti-vAChT) indicated ubiquitous TASK-3 expression in motor neurons, whereas in other brain regions TASK-3 showed a widespread but not ubiquitous expression. In situ hybridization using a TASK-3 specific riboprobe verified the expression of TASK-3 in motor neurons at the mRNA level.


Subject(s)
Motor Neurons/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Immunohistochemistry , Male , Motor Neurons/cytology , Protein Transport , Rats , Rats, Wistar , Spinal Cord/cytology , Spinal Cord/metabolism
12.
Biomolecules ; 12(4)2022 03 25.
Article in English | MEDLINE | ID: mdl-35454090

ABSTRACT

Polyamines (PAs) are small, versatile molecules with two or more nitrogen-containing positively charged groups and provide widespread biological functions. Most of these aspects are well known and covered by quite a number of excellent surveys. Here, the present review includes novel aspects and questions: (1) It summarizes the role of most natural and some important synthetic PAs. (2) It depicts PA uptake from nutrition and bacterial production in the intestinal system following loss of PAs via defecation. (3) It highlights the discrepancy between the high concentrations of PAs in the gut lumen and their low concentration in the blood plasma and cerebrospinal fluid, while concentrations in cellular cytoplasm are much higher. (4) The present review provides a novel and complete scheme for the biosynthesis of Pas, including glycine, glutamate, proline and others as PA precursors, and provides a hypothesis that the agmatine pathway may rescue putrescine production when ODC knockout seems to be lethal (solving the apparent contradiction in the literature). (5) It summarizes novel data on PA transport in brain glial cells explaining why these cells but not neurons preferentially accumulate PAs. (6) Finally, it provides a novel and complete scheme for PA interconversion, including hypusine, putreanine, and GABA (unique gliotransmitter) as end-products. Altogether, this review can serve as an updated contribution to understanding the PA mystery.


Subject(s)
Polyamines , Spermine , Central Nervous System/metabolism , Neuroglia/metabolism , Polyamines/metabolism , Putrescine/metabolism , Spermine/metabolism
13.
Biomolecules ; 12(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36551240

ABSTRACT

The interest in astrocytes, the silent brain cells that accumulate polyamines (PAs), is growing. PAs exert anti-inflammatory, antioxidant, antidepressant, neuroprotective, and other beneficial effects, including increasing longevity in vivo. Unlike neurons, astrocytes are extensively coupled to others via connexin (Cx) gap junctions (GJs). Although there are striking modulatory effects of PAs on neuronal receptors and channels, PA regulation of the astrocytic GJs is not well understood. We studied GJ-propagation using molecules of different (i) electrical charge, (ii) structure, and (iii) molecular weight. Loading single astrocytes with patch pipettes containing membrane-impermeable dyes, we observed that (i) even small molecules do not easily permeate astrocytic GJs, (ii) the ratio of the charge to weight of these molecules is the key determinant of GJ permeation, (iii) the PA spermine (SPM) induced the propagation of negatively charged molecules via GJs, (iv) while no effects were observed on propagation of macromolecules with net-zero charge. The GJ uncoupler carbenoxolone (CBX) blocked such propagation. Taken together, these findings indicate that SPM is essential for astrocytic GJ communication and selectively facilitates intracellular propagation via GJs for negatively charged molecules through glial syncytium.


Subject(s)
Polyamines , Spermine , Spermine/pharmacology , Polyamines/pharmacology , Astrocytes , Gap Junctions , Giant Cells
14.
Glia ; 59(6): 893-902, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21438011

ABSTRACT

Large-conductance Ca(2+) -activated (BK) potassium channels are centrally involved in neurovascular coupling, immunity, and neural transmission. The ability to be synergistically activated by membrane depolarization, different ligands and intracellular Ca(2+) links intracellular signaling and membrane excitability. The diverse physiological functions of BK channels crucially depend on regulatory ß subunits. Although first studies characterized the neuronal distribution of BKß subunits in the rodent brain, it is largely unknown which ß subunit proteins are expressed in astrocytes and thus mediate these regulatory effects. We therefore analyzed the expression of BKß subunits in rat and mouse brain and glial cell cultures. A monospecific polyclonal antibody against the BKß4 channel subunit was raised, affinity-purified and extensively characterized. BKß4 and to a lesser degree BKß1 transcripts and protein were detected in several astrocytic populations and cultured cells. Particularly strong BKß4 immunostaining was detected in astrocytic progenitors derived from the subventricular zone. The overlapping expression of BKα and BKß4 in astrocytes implies a functional relationship and suggests that BKß4 is an important accessory ß subunit for astrocytic BK channels. In addition, BKß4 might exert effects independent of the α subunit as functional heterologous co-expression of Nav1.6 and BKß4 resulted in reduced Nav1.6 sodium currents. Thus, BKß4 expression in astrocytes likely participates in regulating astrocytic voltage gradients and maintaining K(+) homeostasis, hence enabling astrocytes to fulfill their complex regulatory influence on proper brain function.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Large-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Astrocytes/cytology , Brain/cytology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , HEK293 Cells , Homeostasis/genetics , Humans , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Inbred C57BL , Oocytes , Rats , Rats, Wistar , Xenopus laevis
15.
Cell Mol Neurobiol ; 31(2): 323-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21082237

ABSTRACT

Monoaminergic and cholinergic systems are important regulators of cortical and subcortical systems, and a variety of vegetative functions are controlled by the respective neurotransmitters. Neuronal excitability and transmitter release of these neurons are strongly regulated by their potassium conductances carried by Kir and K(2P) channels. Here we describe the generation and characterization of a polyclonal monospecific antibody against rat TASK-3, a major brain K(2P) channel. After removal of cross-reactivities and affinity purification the antibody was characterized by ELISA, immunocytochemistry of TASK-3 transfected cells, and Western blots indicating that the antibody only detects TASK-3 protein, but not its paralogs TASK-1 and TASK-5. Western blot analysis of brain membrane fractions showed a single band around 45 kD, close to the predicted molecular weight of the TASK-3 protein. In addition, specific immunolabeling using the anti-TASK-3 antibody in Western blot analysis and immunocytochemistry was blocked in a concentration dependent manner by its cognate antigen only. Immunocytochemical analysis of rat brain revealed strong expression of TASK-3 channels in serotoninergic neurons of the dorsal and median raphe, noradrenergic neurons of the locus coeruleus, histaminergic neurons of the tuberomammillary nucleus and in the cholinergic neurons of the basal nucleus of Meynert. Immunofluorescence double-labeling experiments with appropriate marker enzymes confirmed the expression of TASK-3 in cholinergic, serotoninergic, and noradrenergic neurons. In the dopaminergic system strong TASK-3 expression was found in the ventral tegmental area, whereas TASK-3 immunoreactivity in the substantia nigra compacta was only weak. All immunocytochemical results were supported by in situ hybridization using TASK-3 specific riboprobes.


Subject(s)
Choline/metabolism , Neurons/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Antibody Specificity/immunology , Basal Nucleus of Meynert/cytology , Basal Nucleus of Meynert/metabolism , Blotting, Western , Brain/metabolism , Dopamine/metabolism , Histamine/metabolism , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Immunohistochemistry , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Male , Neurons/cytology , Norepinephrine/metabolism , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Serotonin/metabolism , Transfection
16.
Anal Bioanal Chem ; 400(5): 1367-73, 2011 May.
Article in English | MEDLINE | ID: mdl-21479547

ABSTRACT

Cell cultures form the basis of most biological assays conducted to assess the cytotoxicity of nanomaterials. Since the molecular environment of nanoparticles exerts influence on their physicochemical properties, it can have an impact on nanotoxicity. Here, toxicity of silica nanoparticles upon delivery by fluid-phase uptake is studied in a 3T3 fibroblast cell line. Based on XTT viability assay, cytotoxicity is shown to be a function of (1) particle concentration and (2) of fetal calf serum (FCS) content in the cell culture medium. Application of dynamic light scattering shows that both parameters affect particle agglomeration. The DLS experiments verify the stability of the nanoparticles in culture medium without FCS over a wide range of particle concentrations. The related toxicity can be mainly accounted for by single silica nanoparticles and small agglomerates. In contrast, agglomeration of silica nanoparticles in all FCS-containing media is observed, resulting in a decrease of the associated toxicity. This result has implications for the evaluation of the cytotoxic potential of silica nanoparticles and possibly also other nanomaterials in standard cell culture.


Subject(s)
Cell Survival/drug effects , Fibroblasts/drug effects , Nanoparticles/toxicity , Serum/metabolism , Silicon Dioxide/toxicity , 3T3 Cells , Adsorption , Animals , Blood Proteins , Cattle , Mice
17.
Biomolecules ; 11(8)2021 08 11.
Article in English | MEDLINE | ID: mdl-34439853

ABSTRACT

Polyamines (PAs) are polycationic biomolecules containing multiple amino groups. Patients with HIV-associated neurocognitive disorder (HAND) have high concentrations of the polyamine N-acetylated spermine in their brain and cerebral spinal fluid (CSF) and have increased PA release from astrocytes. These effects are due to the exposure to HIV-Tat. In healthy adult brain, PAs are accumulated but not synthesized in astrocytes, suggesting that PAs must enter astrocytes to be N-acetylated and released. Therefore, we tested if Cx43 hemichannels (Cx43-HCs) are pathways for PA flux in control and HIV-Tat-treated astrocytes. We used biotinylated spermine (b-SPM) to examine polyamine uptake. We found that control astrocytes and those treated with siRNA-Cx43 took up b-SPM, similarly suggesting that PA uptake is via a transporter/channel other than Cx43-HCs. Surprisingly, astrocytes pretreated with both HIV-Tat and siRNA-Cx43 showed increased accumulation of b-SPM. Using a novel polyamine transport inhibitor (PTI), trimer 44NMe, we blocked b-SPM uptake, showing that PA uptake is via a PTI-sensitive transport mechanism such as organic cation transporter. Our data suggest that Cx43 HCs are not a major pathway for b-SPM uptake in the condition of normal extracellular calcium concentration but may be involved in the release of PAs to the extracellular space during viral infection.


Subject(s)
Astrocytes/metabolism , Biological Transport/drug effects , Connexin 43/metabolism , HIV Infections/metabolism , Spermine/metabolism , Animals , Astrocytes/virology , HIV-1 , Mice , Mice, Inbred C57BL , Primary Cell Culture
18.
Front Cell Neurosci ; 15: 787319, 2021.
Article in English | MEDLINE | ID: mdl-35069115

ABSTRACT

Accumulating evidence indicate that astrocytes are essential players of the excitatory and inhibitory signaling during normal and epileptiform activity via uptake and release of gliotransmitters, ions, and other substances. Polyamines can be regarded as gliotransmitters since they are almost exclusively stored in astrocytes and can be released by various mechanisms. The polyamine putrescine (PUT) is utilized to synthesize GABA, which can also be released from astrocytes and provide tonic inhibition on neurons. The polyamine spermine (SPM), synthesized form PUT through spermidine (SPD), is known to unblock astrocytic Cx43 gap junction channels and therefore facilitate astrocytic synchronization. In addition, SPM released from astrocytes may also modulate neuronal NMDA, AMPA, and kainate receptors. As a consequence, astrocytic polyamines possess the capability to significantly modulate epileptiform activity. In this study, we investigated different steps in polyamine metabolism and coupled GABA release to assess their potential to control seizure generation and maintenance in two different epilepsy models: the low-[Mg2+] model of temporal lobe epilepsy in vitro and in the WAG/Rij rat model of absence epilepsy in vivo. We show that SPM is a gliotransmitter that is released from astrocytes and significantly contributes to network excitation. Importantly, we found that inhibition of SPD synthesis completely prevented seizure generation in WAG/Rij rats. We hypothesize that this antiepileptic effect is attributed to the subsequent enhancement of PUT to GABA conversion in astrocytes, leading to GABA release through GAT-2/3 transporters. This interpretation is supported by the observation that antiepileptic potential of the Food and Drug Administration (FDA)-approved drug levetiracetam can be diminished by specifically blocking astrocytic GAT-2/3 with SNAP-5114, suggesting that levetiracetam exerts its effect by increasing surface expression of GAT-2/3. Our findings conclusively suggest that the major pathway through which astrocytic polyamines contribute to epileptiform activity is the production of GABA. Modulation of astrocytic polyamine levels, therefore, may serve for a more effective antiepileptic drug development in the future.

19.
Pharmacogenet Genomics ; 20(5): 291-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20220551

ABSTRACT

AIM: KCNJ6 coding for potassium inwardly rectifying channels (Kir3.2, GIRK2) is important for opioid receptor transmission. The KCNJ6 rs2070995 AA genotype has been associated with increased opioid analgesic requirements in Japanese. We analyzed its consequences for other opioid effects. METHODS: Genotyping was done in 85 methadone-substituted former heroin addicts, 352 opioid-treated chronic pain patients, and in 51 healthy volunteers where miotic effects of levomethadone had been measured. Expression of Kir3.2 in the Edinger-Westphal nucleus of rat brains was analyzed by means of immunohistochemistry. RESULTS: Average daily methadone substitution doses during the first therapy year were larger in the AA genotype (n=4, 119.7+/-49.6 mg/day) than in other rs2070995 genotypes (77.5+/-26.2 mg/day, P=0.003) whereas AA carriers lacked opioid withdrawal symptoms. A similar tendency toward less opioid effectiveness was observed toward higher opioid dosing demands for analgesia in the AA genotype (n=17, opioid dose 2.03+/-0.45 log mg oral morphine equivalents per day, controls: 1.81+/-0.52 log mg oral morphine equivalents/day, P=0.093). In contrast, no pharmacogenetic effects were observed on miotic opioid effects. This could be traced back to the absence of Kir3.2 from the Edinger-Westphal nucleus in rat brains, a key cerebral structure governing pupil constriction. CONCLUSION: The association of the KCNJ6 rs2070995 AA genotype with increased opioid requirements extends from analgesia to opiate substitution therapy. Opioid induced miosis is exempted for molecular histological reasons.


Subject(s)
Analgesia/methods , Analgesics, Opioid/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Polymorphism, Genetic , Administration, Oral , Animals , Brain/metabolism , Genotype , Heroin/pharmacology , Japan , Methadone/pharmacology , Morphine/pharmacology , Rats , Rats, Wistar , Receptors, Opioid/metabolism
20.
Histochem Cell Biol ; 133(3): 301-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20012645

ABSTRACT

The development of the hippocampal network requires neuronal activity, which is shaped by the differential expression and sorting of a variety of potassium channels. Parallel to their maturation, hippocampal neurons undergo a distinct development of their ion channel profile. The age-dependent dimension of ion channel occurrence is of utmost importance as it is interdependently linked to network formation. However, data regarding the exact temporal expression of potassium channels during postnatal hippocampal development are scarce. We therefore studied the expression of several voltage-gated potassium channel proteins during hippocampal development in vivo and in primary cultures, focusing on channels that were sorted to the axonal compartment. The Kv1.1, Kv1.2, Kv1.4, and Kv3.4 proteins showed a considerable temporal variation of axonal localization among neuronal subpopulations. It is possible, therefore, that hippocampal neurons possess cell type-specific mechanisms for channel compartmentalization. Thus, age-dependent axonal sorting of the potassium channel proteins offers a new approach to functionally distinguish classes of hippocampal neurons and may extend our understanding of hippocampal circuitry and memory processing.


Subject(s)
Aging/physiology , Axons/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism , Animals , Cell Culture Techniques , Hippocampus/metabolism , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/biosynthesis , Potassium Channels/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL