Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047724

ABSTRACT

The analysis of histological alterations in all types of tissue is of primary importance in pathology for highly accurate and robust diagnosis. Recent advances in tissue clearing and fluorescence microscopy made the study of the anatomy of biological tissue possible in three dimensions. The combination of these techniques with classical hematoxylin and eosin (H&E) staining has led to the birth of three-dimensional (3D) histology. Here, we present an overview of the state-of-the-art methods, highlighting the optimal combinations of different clearing methods and advanced fluorescence microscopy techniques for the investigation of all types of biological tissues. We employed fluorescence nuclear and eosin Y staining that enabled us to obtain hematoxylin and eosin pseudo-coloring comparable with the gold standard H&E analysis. The computational reconstructions obtained with 3D optical imaging can be analyzed by a pathologist without any specific training in volumetric microscopy, paving the way for new biomedical applications in clinical pathology.


Subject(s)
Imaging, Three-Dimensional , Hematoxylin , Eosine Yellowish-(YS) , Microscopy, Fluorescence/methods , Staining and Labeling , Imaging, Three-Dimensional/methods , Microscopy, Confocal
2.
Life Sci Alliance ; 7(1)2024 01.
Article in English | MEDLINE | ID: mdl-37923359

ABSTRACT

The hERG1 potassium channel is aberrantly over expressed in tumors and regulates the cancer cell response to integrin-dependent adhesion. We unravel a novel signaling pathway by which integrin engagement by the ECM protein fibronectin promotes hERG1 translocation to the plasma membrane and its association with ß1 integrins, by activating girdin-dependent Gαi3 proteins and protein kinase B (Akt). By sequestering hERG1, ß1 integrins make it avoid Rab5-mediated endocytosis, where unbound channels are degraded. The cycle of hERG1 expression determines the resting potential (Vrest) oscillations and drives the cortical f-actin dynamics and thus cell motility. To interpret the slow biphasic kinetics of hERG1/ß1 integrin interplay, we developed a mathematical model based on a generic balanced inactivation-like module. Integrin-mediated cell adhesion triggers two contrary responses: a rapid stimulation of hERG1/ß1 complex formation, followed by a slow inhibition which restores the initial condition. The protracted hERG1/ß1 integrin cycle determines the slow time course and cyclic behavior of cell migration in cancer cells.


Subject(s)
Integrins , Neoplasms , Humans , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Integrin beta1/metabolism , Integrins/metabolism , Neoplasms/pathology , Signal Transduction
3.
Heliyon ; 9(10): e20112, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37767500

ABSTRACT

Pharmacological studies aimed at the development of newly synthesized drugs directed against ion channels (as well as genetic studies of ion channel mutations) involve the development and use of transfected cells. However, the identification of the best clone, in terms of transfection efficiency, is often a time consuming procedure when performed through traditional methods such as manual patch-clamp. On the other hand, the use of other faster techniques, such as for example the IF, are not informative on the effective biological functionality of the transfected ion channel(s). In the present work, we used the high throughput automated ion channel reader (ICR) technology (ICR8000 Aurora Biomed Inc.) that combine atomic absorption spectroscopy with a patented microsampling process to accurately measure ion flux in cell-based screening assays. This technology indeed helped us to evaluate the transfection efficiency of hERG1 and hKv1.3 channels respectively on the HEK-293 and CHO cellular models. Moreover, as proof of the validity of this innovative method, we have corroborated these data with the functional characterization of the potassium currents carried out by the same clones through patch-clamp recordings. The results obtained in our study are promising and represent a valid methodological strategy to screen a large number of clones simultaneously and to pharmacologically evaluate their functionality within an extremely faster timeframe.

4.
Eur J Med Chem ; 259: 115561, 2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37454520

ABSTRACT

Voltage-gated potassium channel KV1.3 inhibitors have been shown to be effective in preventing T-cell proliferation and activation by affecting intracellular Ca2+ homeostasis. Here, we present the structure-activity relationship, KV1.3 inhibition, and immunosuppressive effects of new thiophene-based KV1.3 inhibitors with nanomolar potency on K+ current in T-lymphocytes and KV1.3 inhibition on Ltk- cells. The new KV1.3 inhibitor trans-18 inhibited KV1.3 -mediated current in phytohemagglutinin (PHA)-activated T-lymphocytes with an IC50 value of 26.1 nM and in mammalian Ltk- cells with an IC50 value of 230 nM. The KV1.3 inhibitor trans-18 also had nanomolar potency against KV1.3 in Xenopus laevis oocytes (IC50 = 136 nM). The novel thiophene-based KV1.3 inhibitors impaired intracellular Ca2+ signaling as well as T-cell activation, proliferation, and colony formation.


Subject(s)
Immunosuppressive Agents , Potassium Channels, Voltage-Gated , Thiophenes , Animals , Mammals/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Potassium Channels/pharmacology , Potassium Channels, Voltage-Gated/pharmacology , Structure-Activity Relationship , T-Lymphocytes , Thiophenes/chemistry , Thiophenes/pharmacology , Immunosuppressive Agents/chemistry
5.
Stem Cells ; 29(12): 2062-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22038821

ABSTRACT

Adult spinal cord has little regenerative potential, thus limiting patient recovery following injury. In this study, we describe a new population of cells resident in the adult rat spinal cord meninges that express the neural stem/precursor markers nestin and doublecortin. Furthermore, from dissociated meningeal tissue a neural stem cell population was cultured in vitro and subsequently shown to differentiate into functional neurons or mature oligodendrocytes. Proliferation rate and number of nestin- and doublecortin-positive cells increased in vivo in meninges following spinal cord injury. By using a lentivirus-labeling approach, we show that meningeal cells, including nestin- and doublecortin-positive cells, migrate in the spinal cord parenchyma and contribute to the glial scar formation. Our data emphasize the multiple roles of meninges in the reaction of the parenchyma to trauma and indicate for the first time that spinal cord meninges are potential niches harboring stem/precursor cells that can be activated by injury. Meninges may be considered as a new source of adult stem/precursor cells to be further tested for use in regenerative medicine applied to neurological disorders, including repair from spinal cord injury.


Subject(s)
Intermediate Filament Proteins/metabolism , Meninges/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Spinal Cord Injuries/therapy , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Doublecortin Domain Proteins , Doublecortin Protein , Electrophysiologic Techniques, Cardiac , Gene Expression Profiling , Intermediate Filament Proteins/genetics , Laminectomy , Lentivirus/genetics , Lentivirus/metabolism , Meninges/cytology , Meninges/physiology , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Nestin , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Neuropeptides/genetics , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Regenerative Medicine , Stem Cell Niche
6.
Membranes (Basel) ; 12(11)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36422154

ABSTRACT

Ion channels are implicated in various diseases, including cancer, in which they modulate different aspects of cancer progression. In particular, potassium channels are often aberrantly expressed in cancers, a major example being provided by hERG1. The latter is generally complexed with ß1 integrin in tumour cells, and such a molecular complex represents a new druggable hub. The present study focuses on the characterization of the functional consequences of the interaction between hERG1 and ß1 integrins on different substrates over time. To this purpose, we studied the interplay alteration on the plasma membrane through patch clamp techniques in a cellular model consisting of human embryonic kidney (HEK) cells stably transfected with hERG1 and in a cancer cell model consisting of SH-SY5Y neuroblastoma cells, endogenously expressing the channel. Cells were seeded on different substrates known to stimulate ß1 integrins, such as fibronectin (FN) for HEK-hERG1 and laminin (LMN) for SH-SY5Y. In HEK cells stably overexpressing hERG1, we observed a hERG1 current density increase accompanied by Vrest hyperpolarization after cell seeding onto FN. Notably, a similar behaviour was shown by SH-SY5Y neuroblastoma cells plated onto LMN. Interestingly, we did not observe this phenomenon when plating the cells on substrates such as Bovine Serum Albumin (BSA) or Polylysine (PL), thus suggesting a crucial involvement of ECM proteins as well as of ß1 integrin activation.

7.
Data Brief ; 34: 106668, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33385031

ABSTRACT

In the present work, applying the whole-cell patch-clamp technique in voltage clamp mode, we have investigated the effects of different drugs, such as riluzole, Psora-4 and Tram-34, on the potassium currents in four human lymphoma cell lines. We focused on outward currents mediated by two potassium channels (Kv1.3 and KCa3.1), which are known to play a key physiological role in lymphoid cells. The currents were evoked by voltage ramps ranging from -120 mV to +40 mV and the conductance of the two potassium channels was measured between +20 mV and +40 mV, both in the absence and in the presence of the specific blockers Psora-4 (Kv1.3; 1 µM) and Tram-34 (KCa3.1; 1 µM). The effect of the latter was tested after KCa3.1 channels were activated by riluzole 10 µM. Taken together, these data could be useful as an indication of the functional characteristics of the potassium channels in human lymphomas and represent a starting point for the study of potassium conductance in cellular models of these tumors.

8.
PLoS One ; 14(9): e0222855, 2019.
Article in English | MEDLINE | ID: mdl-31557210

ABSTRACT

Trazodone is an antidepressant drug with considerable affinity for 5-HT1A receptors and α1-adrenoceptors for which the drug is competitive agonist and antagonist, respectively. In this study, we used cell-attached or whole-cell patch-clamp recordings to characterize the effects of trazodone at somatodendritic 5-HT1A receptors (5-HT1AARs) and α1-adrenoceptors of serotonergic neurons in rodent dorsal raphe slices. To reveal the effects of trazodone at α1-adrenoceptors, the baseline firing of 5-HT neurons was facilitated by applying the selective α1-adrenoceptor agonist phenylephrine at various concentrations. In the absence of phenylephrine, trazodone (1-10 µM) concentration-dependently silenced neurons through activation of 5-HT1AARs. The effect was fully antagonized by the selective 5-HT1A receptor antagonist Way-100635. With 5-HT1A receptors blocked by Way-100635, trazodone (1-10 µM) concentration-dependently inhibited neuron firing facilitated by 1 µM phenylephrine. Parallel rightward shift of dose-response curves for trazodone recorded in higher phenylephrine concentrations (10-100 µM) indicated competitive antagonism at α1-adrenoceptors. Both effects of trazodone were also observed in slices from Tph2-/- mice that lack synthesis of brain serotonin, showing that the activation of 5-HT1AARs was not mediated by endogenous serotonin. In whole-cell recordings, trazodone activated 5-HT1AAR-coupled G protein-activated inwardly-rectifying (GIRK) channel conductance with weak partial agonist efficacy (~35%) compared to that of the full agonist 5-CT. Collectively our data show that trazodone, at concentrations relevant to its clinical effects, exerts weak partial agonism at 5-HT1AARs and disfacilitation of firing through α1-adrenoceptor antagonism. These two actions converge in inhibiting dorsal raphe serotonergic neuron activity, albeit with varying contribution depending on the intensity of α1-adrenoceptor stimulation.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Dorsal Raphe Nucleus/drug effects , Serotonergic Neurons/drug effects , Serotonin 5-HT1 Receptor Agonists/pharmacology , Trazodone/pharmacology , Animals , Dorsal Raphe Nucleus/cytology , Dorsal Raphe Nucleus/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Knockout , Patch-Clamp Techniques , Phenylephrine/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Serotonergic Neurons/metabolism , Tryptophan Hydroxylase/genetics
9.
J Biomed Opt ; 24(10): 1-6, 2019 10.
Article in English | MEDLINE | ID: mdl-31674164

ABSTRACT

Confocal detection in digital scanned laser light-sheet fluorescence microscopy (DSLM) has been established as a gold standard method to improve image quality. The selective line detection of a complementary metal­oxide­semiconductor camera (CMOS) working in rolling shutter mode allows the rejection of out-of-focus and scattered light, thus reducing background signal during image formation. Most modern CMOS have two rolling shutters, but usually only a single illuminating beam is used, halving the maximum obtainable frame rate. We report on the capability to recover the full image acquisition rate via dual confocal DSLM by using an acousto-optic deflector. Such a simple solution enables us to independently generate, control and synchronize two beams with the two rolling slits on the camera. We show that the doubling of the imaging speed does not affect the confocal detection high contrast.


Subject(s)
Image Processing, Computer-Assisted/methods , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Animals , Brain/diagnostic imaging , Equipment Design , High-Throughput Screening Assays/methods , Larva/cytology , Mice , Mice, Inbred C57BL , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence/instrumentation , Zebrafish
10.
Front Neurosci ; 13: 245, 2019.
Article in English | MEDLINE | ID: mdl-31068767

ABSTRACT

Brain serotonin (5-hydroxytryptamine, 5-HT) system dysfunction is implicated in exaggerated fear responses triggering various anxiety-, stress-, and trauma-related disorders. However, the underlying mechanisms are not well understood. Here, we investigated the impact of constitutively inactivated 5-HT synthesis on context-dependent fear learning and extinction using tryptophan hydroxylase 2 (Tph2) knockout mice. Fear conditioning and context-dependent fear memory extinction paradigms were combined with c-Fos imaging and electrophysiological recordings in the dorsal hippocampus (dHip). Tph2 mutant mice, completely devoid of 5-HT synthesis in brain, displayed accelerated fear memory formation and increased locomotor responses to foot shock. Furthermore, recall of context-dependent fear memory was increased. The behavioral responses were associated with increased c-Fos expression in the dHip and resistance to foot shock-induced impairment of hippocampal long-term potentiation (LTP). In conclusion, increased context-dependent fear memory resulting from brain 5-HT deficiency involves dysfunction of the hippocampal circuitry controlling contextual representation of fear-related behavioral responses.

11.
Eur Neuropsychopharmacol ; 27(12): 1258-1267, 2017 12.
Article in English | MEDLINE | ID: mdl-29126768

ABSTRACT

Firing activity of serotonergic neurons is under regulatory control by somatodendritic 5-HT1A autoreceptors (5-HT1AARs). Enhanced 5-HT1AAR functioning may cause decreased serotonergic signaling in brain and has thereby been implicated in the etiology of mood and anxiety disorders. Tryptophan hydroxylase-2 knockout (Tph2-/-) mice exhibit sensitization of 5-HT1A agonist-induced inhibition of serotonergic neuron firing and thus represents a unique animal model of enhanced 5-HT1AAR functioning. To elucidate the mechanisms underlying 5-HT1AAR supersensitivity in Tph2-/- mice, we characterized the activation of G protein-coupled inwardly-rectifying potassium (GIRK) conductance by the 5-HT1A receptor agonist 5-carboxamidotryptamine using whole-cell recordings from serotonergic neurons in dorsal raphe nucleus. Tph2-/- mice exhibited a mean twofold leftward shift of the agonist concentration-response curve (p < 0.001) whereas the maximal response, proportional to the 5-HT1AAR number, was not different (p = 0.42) compared to Tph2+/- and Tph2+/+ littermates. No differences were found in the basal inwardly-rectifying potassium conductance, determined in the absence of agonist, (p = 0.80) nor in total GIRK conductance activated by intracellular application of GTP-γ-S (p = 0.69). These findings indicate increased functional coupling of 5-HT1AARs to GIRK channels in Tph2-/- mice without a concomitant increase in 5-HT1AARs and/or GIRK channel density. In addition, no changes were found in α1-adrenergic facilitation of firing (p = 0.72) indicating lack of adaptive changes Tph2-/- mice. 5-HT1AAR supersensitivity may represents a previously unrecognized cause of serotonergic system hypofunction and associated disorders and provides a possible explanation for conflicting results on the correlation between 5-HT1AAR density and depression in clinical imaging studies.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Regulation/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Serotonergic Neurons/physiology , Tryptophan Hydroxylase/deficiency , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Dorsal Raphe Nucleus/cytology , Dose-Response Relationship, Drug , Electric Stimulation , Female , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , In Vitro Techniques , Male , Mice , Mice, Knockout , Patch-Clamp Techniques , Phosphinic Acids/pharmacology , Propanolamines/pharmacology , Serotonergic Neurons/drug effects , Serotonin/analogs & derivatives , Serotonin/pharmacology , Serotonin Agents/pharmacology , Time Factors , Tryptophan Hydroxylase/genetics
12.
Front Cell Neurosci ; 10: 195, 2016.
Article in English | MEDLINE | ID: mdl-27536220

ABSTRACT

Tonic spiking of serotonergic neurons establishes serotonin levels in the brain. Since the first observations, slow regular spiking has been considered as a defining feature of serotonergic neurons. Recent studies, however, have revealed the heterogeneity of serotonergic neurons at multiple levels, comprising their electrophysiological properties, suggesting the existence of functionally distinct cellular subpopulations. In order to examine in an unbiased manner whether serotonergic neurons of the dorsal raphe nucleus (DRN) are heterogeneous, we used a non-invasive loose-seal cell-attached method to record α1 adrenergic receptor-stimulated spiking of a large sample of neurons in brain slices obtained from transgenic mice lines that express fluorescent marker proteins under the control of serotonergic system-specific Tph2 and Pet-1 promoters. We found wide homogeneous distribution of firing rates, well fitted by a single Gaussian function (r (2) = 0.93) and independent of anatomical location (P = 0.45), suggesting that in terms of intrinsic firing properties, serotonergic neurons in the DRN represent a single cellular population. Characterization of the population in terms of spiking regularity was hindered by its dependence on the firing rate. For instance, the coefficient of variation of the interspike intervals (ISI), a common measure of spiking irregularity, is of limited usefulness since it correlates negatively with the firing rate (r = -0.33, P < 0.0001). Nevertheless, the majority of neurons exhibited regular, pacemaker-like activity, with coefficient of variance of the ISI lower than 0.5 in ~97% of cases. Unexpectedly, a small percentage of neurons (~1%) exhibited a particular spiking pattern, characterized by low frequency (~0.02-0.1 Hz) oscillations in the firing rate. Transitions between regular and oscillatory firing were observed, suggesting that the oscillatory firing is an alternative firing pattern of serotonergic neurons.

13.
PLoS One ; 10(10): e0140369, 2015.
Article in English | MEDLINE | ID: mdl-26460748

ABSTRACT

G protein-activated inwardly rectifying potassium (GIRK) channels in 5-HT neurons are assumed to be principal effectors of 5-hydroxytryptamine 1A (5-HT1A) autoreceptors, but their pharmacology, subunit composition and the role in regulation of 5-HT neuron activity have not been fully elucidated. We sought for a pharmacological tool for assessing the functional role of GIRK channels in 5-HT neurons by characterizing the effects of drugs known to block GIRK channels in the submicromolar range of concentrations. Whole-cell voltage-clamp recording in brainstem slices were used to determine concentration-response relationships for the selected GIRK channel blockers on 5-HT1A autoreceptor-activated inwardly rectifying K+ conductance in rat dorsal raphe 5-HT neurons. 5-HT1A autoreceptor-activated GIRK conductance was completely blocked by the nonselective inwardly rectifying potassium channels blocker Ba2+ (EC50 = 9.4 µM, full block with 100 µM) and by SCH23390 (EC50 = 1.95 µM, full block with 30 µM). GIRK-specific blocker tertiapin-Q blocked 5-HT1A autoreceptor-activated GIRK conductance with high potency (EC50 = 33.6 nM), but incompletely, i.e. ~16% of total conductance resulted to be tertiapin-Q-resistant. U73343 and SCH28080, reported to block GIRK channels with submicromolar EC50s, were essentially ineffective in 5-HT neurons. Our data show that inwardly rectifying K+ channels coupled to 5-HT1A autoreceptors display pharmacological properties generally expected for neuronal GIRK channels, but different from GIRK1-GIRK2 heteromers, the predominant form of brain GIRK channels. Distinct pharmacological properties of GIRK channels in 5-HT neurons should be explored for the development of new therapeutic agents for mood disorders.


Subject(s)
Dorsal Raphe Nucleus/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonergic Neurons/metabolism , Animals , Barium/pharmacology , Bee Venoms/pharmacology , Benzazepines/pharmacology , Dorsal Raphe Nucleus/drug effects , Electric Conductivity , Estrenes/pharmacology , Male , Maleates/pharmacology , Pyrrolidinones/pharmacology , Rats, Wistar , Serotonergic Neurons/drug effects
14.
Eur Neuropsychopharmacol ; 25(11): 2022-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26409296

ABSTRACT

Considerable evidence links dysfunction of serotonin (5-hydroxytryptamine, 5-HT) transmission to neurodevelopmental and psychiatric disorders characterized by compromised "social" cognition and emotion regulation. It is well established that the brain 5-HT system is under autoregulatory control by its principal transmitter 5-HT via its effects on activity and expression of 5-HT system-related proteins. To examine whether 5-HT itself also has a crucial role in the acquisition and maintenance of characteristic rhythmic firing of 5-HT neurons, we compared their intrinsic electrophysiological properties in mice lacking brain 5-HT, i.e. tryptophan hydroxylase-2 null mice (Tph2(-/-)) and their littermates, Tph2(+/-) and Tph2(+/+), by using whole-cell patch-clamp recordings in a brainstem slice preparation and single unit recording in anesthetized animals. We report that the active properties of dorsal raphe nucleus (DRN) 5-HT neurons in vivo (firing rate magnitude and variability; the presence of spike doublets) and in vitro (firing in response to depolarizing current pulses; action potential shape) as well as the resting membrane potential remained essentially unchanged across Tph2 genotypes. However, there were subtle differences in subthreshold properties, most notably, an approximately 25% higher input conductance in Tph2(-/-) mice compared with Tph2(+/-) and Tph2(+/+) littermates (p<0.0001). This difference may at least in part be a consequence of slightly bigger size of the DRN 5-HT neurons in Tph2(-/-) mice (approximately 10%, p<0.0001). Taken together, these findings show that 5-HT neurons acquire and maintain their signature firing properties independently of the presence of their principal neurotransmitter 5-HT, displaying an unexpected functional resilience to complete brain 5-HT deficiency.


Subject(s)
Action Potentials/physiology , Dorsal Raphe Nucleus/physiology , Serotonergic Neurons/physiology , Tryptophan Hydroxylase/deficiency , Action Potentials/drug effects , Animals , Dorsal Raphe Nucleus/cytology , Dorsal Raphe Nucleus/drug effects , Electric Capacitance , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice, Knockout , Microscopy, Fluorescence , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels/metabolism , Serotonergic Neurons/cytology , Serotonergic Neurons/drug effects , Tissue Culture Techniques , Tryptophan Hydroxylase/genetics
15.
J Gen Physiol ; 145(3): 225-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25712017

ABSTRACT

The firing activity of serotonergic neurons in raphe nuclei is regulated by negative feedback exerted by extracellular serotonin (5-HT)o acting through somatodendritic 5-HT1A autoreceptors. The steady-state [5-HT]o, sensed by 5-HT1A autoreceptors, is determined by the balance between the rates of 5-HT release and reuptake. Although it is well established that reuptake of 5-HTo is mediated by 5-HT transporters (SERT), the release mechanism has remained unclear. It is also unclear how selective 5-HT reuptake inhibitor (SSRI) antidepressants increase the [5-HT]o in raphe nuclei and suppress serotonergic neuron activity, thereby potentially diminishing their own therapeutic effect. Using an electrophysiological approach in a slice preparation, we show that, in the dorsal raphe nucleus (DRN), continuous nonexocytotic 5-HT release is responsible for suppression of phenylephrine-facilitated serotonergic neuron firing under basal conditions as well as for autoinhibition induced by SSRI application. By using 5-HT1A autoreceptor-activated G protein-gated inwardly rectifying potassium channels of patched serotonergic neurons as 5-HTo sensors, we show substantial nonexocytotic 5-HT release under conditions of abolished firing activity, Ca(2+) influx, vesicular monoamine transporter 2-mediated vesicular accumulation of 5-HT, and SERT-mediated 5-HT transport. Our results reveal a cytosolic origin of 5-HTo in the DRN and suggest that 5-HTo may be supplied by simple diffusion across the plasma membrane, primarily from the dense network of neurites of serotonergic neurons surrounding the cell bodies. These findings indicate that the serotonergic system does not function as a sum of independently acting neurons but as a highly interdependent neuronal network, characterized by a shared neurotransmitter pool and the regulation of firing activity by an interneuronal, yet activity-independent, nonexocytotic mechanism.


Subject(s)
Action Potentials , Exocytosis , Serotonergic Neurons/metabolism , Serotonin/metabolism , Animals , Calcium/metabolism , Cells, Cultured , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Rats , Rats, Wistar , Serotonergic Neurons/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/metabolism
16.
Front Cell Neurosci ; 8: 18, 2014.
Article in English | MEDLINE | ID: mdl-24550777

ABSTRACT

Rett Syndrome (RTT) is a neurodevelopmental disorder associated with intellectual disability, mainly caused by loss-of-function mutations in the MECP2 gene. RTT brains display decreased neuronal size and dendritic arborization possibly caused by either a developmental failure or a deficit in the maintenance of dendritic arbor structure. To distinguish between these two hypotheses, the development of Mecp2-knockout mouse hippocampal neurons was analyzed in vitro. Since a staging system for the in vitro development of mouse neurons was lacking, mouse and rat hippocampal neurons development was compared between 1-15 days in vitro (DIV) leading to a 6-stage model for both species. Mecp2-knockout hippocampal neurons displayed reduced growth of dendritic branches from stage 4 (DIV4) onwards. At stages 5-6 (DIV9-15), synapse number was lowered in Mecp2-knockout neurons, suggesting increased synapse elimination. These results point to both a developmental and a maintenance setback affecting the final shape and function of neurons in RTT.

SELECTION OF CITATIONS
SEARCH DETAIL